1
|
Li XL, Li M, Yang H, Tian J, Shi ZW, Wang LZ, Song K. Chronic myelogenous leukemia secondary to colon cancer: A case report. World J Gastrointest Oncol 2025; 17:102021. [DOI: 10.4251/wjgo.v17.i4.102021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/26/2025] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Colon cancer is a common malignancy of the digestive tract. An estimated 1148515 new cases of colon cancer were reported in 2020 worldwide. Chronic myeloid leukemia (CML) is a malignant tumor formed by the clonal proliferation of bone marrow hematopoietic stem cells, with an annual incidence rate of 1-2 cases per 100000 people worldwide. Leukemia can be secondary to solid tumors, and vice versa. Reports on CML secondary malignant tumors account for 8.7% but CML secondary to malignancy is extremely rare. Therapy-related CML is a rare but potentially fatal adverse event of chemotherapy or radiotherapy. Herein, we report a case of CML with colon cancer and discuss this unique patient population. Our findings can provide effective raw data and guidance for the diagnosis of this clinical disease.
CASE SUMMARY A 61-year-old male patient attended our hospital due to leukocytosis for 5 days. In February 2020, the patient was diagnosed with colon cancer and underwent radical surgery and conventional chemotherapy with a stable condition. He was diagnosis was CML-chronic phase (Sokal score of 0.72) after examination. He was treated with imatinib (400 mg daily). When his conditions improved, the patient was discharged from our hospital and visited the outpatient department for follow-up twice a month.
CONCLUSION CML in patients with colon cancer is extremely rare. Secondary hematological tumors may be multifactorial, and the exact mechanism is currently unknown. Owing to the slow progression of the disease, patients with CML show no symptoms in the early stage. However, with disease progression, obvious but non-specific symptoms may appear, including fever, anemia, bleeding tendency, and hypertrophy. Therefore, complete blood count monitoring for routine examination is recommended after cancer treatment for early detection of occult hematological tumors.
Collapse
Affiliation(s)
- Xiao-Lan Li
- Department of Hematology, The First Affiliated Hospital of Jishou University, Jishou 416000, Hunan Province, China
| | - Min Li
- Department of Pharmacy, The First Affiliated Hospital of Jishou University, Jishou 416000, Hunan Province, China
| | - Hua Yang
- Department of Hematology, The First Affiliated Hospital of Jishou University, Jishou 416000, Hunan Province, China
| | - Juan Tian
- Department of Hematology, The First Affiliated Hospital of Jishou University, Jishou 416000, Hunan Province, China
| | - Zi-Wei Shi
- Department of Hematology, The First Affiliated Hospital of Jishou University, Jishou 416000, Hunan Province, China
| | - Ling-Zhi Wang
- Department of Pharmacy, The First Affiliated Hospital of Jishou University, Jishou 416000, Hunan Province, China
| | - Kui Song
- Department of Hematology, The First Affiliated Hospital of Jishou University, Jishou 416000, Hunan Province, China
| |
Collapse
|
2
|
Mahmoud A, Choi PH, Sukhwa C, Pintar J, Walch H, Zhao N, Bermeo J, Chung S, Raghavan M, Bapat S, Jiang Q, Karagkounis G, Meredith J, Giarrizzo M, Firat C, Cercek A, Foote MB, Schultz N, Chatila WK, Nash GM, Shia J, Sanchez-Vega F, Larson S, Dar AC, Rosen N, Ganesh K. Paired primary-metastasis patient-derived organoids and mouse models identify phenotypic evolution and druggable dependencies of peritoneal metastasis from appendiceal cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638725. [PMID: 40027618 PMCID: PMC11870485 DOI: 10.1101/2025.02.17.638725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Peritoneal carcinomatosis is a common yet deadly manifestation of gastrointestinal cancers, with few effective treatments. To identify targetable determinants of peritoneal metastasis, we focused on appendiceal adenocarcinoma (AC), a gastrointestinal cancer that metastasizes almost exclusively to the peritoneum. Current treatments are extrapolated from colorectal cancer (CRC), yet AC has distinct genomic alterations, mucinous morphology and peritoneum restricted metastatic pattern. Further, no stable preclinical models of AC exist, limiting drug discovery and representing an unmet clinical need. We establish a first-in-class stable biobank of 16 long-term cultured AC patient-derived organoids (PDOs), including 3 matched, simultaneously resected primary AC-peritoneal carcinomatosis (AC-PC) pairs. By enriching for cancer cells, AC PDOs enable accurate genomic characterization relative to paucicellular AC tissue. We establish an organoid orthotopic intraperitoneal xenograft model that recapitulates diffuse peritoneal carcinomatosis and show that PC-organoids retain increased metastatic capacity, decreased growth factor dependency and sensitivity to standard of care chemotherapy relative to matched primary AC organoids. Single cell profiling of AC-PC pairs reveals dedifferentiation from mucinous differentiated states in primary AC into intestinal stem cell and fetal progenitor states in AC-PC, with upregulation of oncogenic signaling pathways. Through hypothesis-driven drug testing, we identify KRAS MULTI -ON inhibitor RMC-7977 and Wnt-targeting tyrosine kinase inhibitor WNTinib as novel, clinically actionable strategies to target AC-PC more effectively.
Collapse
|
3
|
Alexander HR, Devi-Chou V. Hepatic Perfusion for Diffuse Metastatic Cancer to the Liver: Open and Percutaneous Techniques. Hematol Oncol Clin North Am 2025; 39:177-190. [PMID: 39510672 DOI: 10.1016/j.hoc.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
The management of patients with diffuse liver metastases remains a significant clinical challenge. In many cancer patients, metastatic disease may be isolated to the liver or the liver may be the dominant site of progressive metastatic cancer. In this setting, progression of disease in the liver generally is the most significant cause of morbidity and mortality.
Collapse
Affiliation(s)
- H Richard Alexander
- Department of Surgery, Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 195 Little Albany Street, Room 2009, New Brunswick, NJ 08901, USA.
| | - Virginia Devi-Chou
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
4
|
Li Y, Cheng Z, Li S, Zhang J. Immunotherapy in colorectal cancer: Statuses and strategies. Heliyon 2025; 11:e41354. [PMID: 39811287 PMCID: PMC11731577 DOI: 10.1016/j.heliyon.2024.e41354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 12/10/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Colorectal cancer (CRC) is widely recognized as the third most prevalent malignancy globally and the second leading cause of cancer-related mortality. Traditional treatment modalities for CRC, including surgery, chemotherapy, and radiotherapy, can be utilized either individually or in combination. However, these treatments frequently result in significant side effects due to their non-specificity and cytotoxicity affecting all cells. Moreover, a considerable number of patients face relapses following these treatments. Consequently, it is imperative to explore more efficacious treatment interventions for CRC patients. Immunotherapy, an emerging frontier in oncology, represents a novel therapeutic approach that leverages the body's immune system to target cancer cells. The principal advantage of immunotherapy is its capacity to selectively target cancer cells while minimizing damage to healthy cells. Its recent adoption as a neoadjuvant therapy presents significant potential to transform the treatment landscape for both primary resectable and metastatic CRC. This review endeavors to offer a comprehensive overview of current strategies in CRC immunotherapy, critically analyze existing literature, underscore anticipated outcomes from ongoing clinical trials, and deliberate on the challenges and impediments encountered within the field of immunotherapy.
Collapse
Affiliation(s)
- Yuan Li
- Shanghai Key Laboratory of Compound Chinese Medicines, The MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zewei Cheng
- Shanghai Key Laboratory of Compound Chinese Medicines, The MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shengli Li
- Precision Research Center for Refractory Diseases and Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Jiwei Zhang
- Shanghai Key Laboratory of Compound Chinese Medicines, The MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
5
|
Zhan T, Betge J, Schulte N, Dreikhausen L, Hirth M, Li M, Weidner P, Leipertz A, Teufel A, Ebert MP. Digestive cancers: mechanisms, therapeutics and management. Signal Transduct Target Ther 2025; 10:24. [PMID: 39809756 PMCID: PMC11733248 DOI: 10.1038/s41392-024-02097-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Cancers of the digestive system are major contributors to global cancer-associated morbidity and mortality, accounting for 35% of annual cases of cancer deaths. The etiologies, molecular features, and therapeutic management of these cancer entities are highly heterogeneous and complex. Over the last decade, genomic and functional studies have provided unprecedented insights into the biology of digestive cancers, identifying genetic drivers of tumor progression and key interaction points of tumor cells with the immune system. This knowledge is continuously translated into novel treatment concepts and targets, which are dynamically reshaping the therapeutic landscape of these tumors. In this review, we provide a concise overview of the etiology and molecular pathology of the six most common cancers of the digestive system, including esophageal, gastric, biliary tract, pancreatic, hepatocellular, and colorectal cancers. We comprehensively describe the current stage-dependent pharmacological management of these malignancies, including chemo-, targeted, and immunotherapy. For each cancer entity, we provide an overview of recent therapeutic advancements and research progress. Finally, we describe how novel insights into tumor heterogeneity and immune evasion deepen our understanding of therapy resistance and provide an outlook on innovative therapeutic strategies that will shape the future management of digestive cancers, including CAR-T cell therapy, novel antibody-drug conjugates and targeted therapies.
Collapse
Affiliation(s)
- Tianzuo Zhan
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johannes Betge
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Schulte
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Dreikhausen
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Hirth
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Moying Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philip Weidner
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Antonia Leipertz
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Teufel
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany.
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
6
|
Murshed I, Bunjo Z, Seow W, Murshed I, Bedrikovetski S, Thomas M, Sammour T. Economic Evaluation of 'Watch and Wait' Following Neoadjuvant Therapy in Locally Advanced Rectal Cancer: A Systematic Review. Ann Surg Oncol 2025; 32:137-157. [PMID: 39181996 PMCID: PMC11659367 DOI: 10.1245/s10434-024-16056-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Owing to multimodal treatment and complex surgery, locally advanced rectal cancer (LARC) exerts a large healthcare burden. Watch and wait (W&W) may be cost saving by removing the need for surgery and inpatient care. This systematic review seeks to identify the economic impact of W&W, compared with standard care, in patients achieving a complete clinical response (cCR) following neoadjuvant therapy for LARC. METHODS The PubMed, OVID Medline, OVID Embase, and Cochrane CENTRAL databases were systematically searched from inception to 26 April 2024. All economic evaluations (EEs) that compared W&W with standard care were included. Reporting and methodological quality was assessed using the Consolidated Health Economic Evaluation Reporting Standards (CHEERS), BMJ and Philips checklists. Narrative synthesis was performed. Primary and secondary outcomes were (incremental) cost-effectiveness ratios and the net financial cost. RESULTS Of 1548 studies identified, 27 were assessed for full-text eligibility and 12 studies from eight countries (2016-2024) were included. Seven cost-effectiveness analyses (complete EEs) and five cost analyses (partial EEs) utilized model-based (n = 7) or trial-based (n = 5) analytics with significant variations in methodological design and reporting quality. W&W showed consistent cost effectiveness (n = 7) and cost saving (n = 12) compared with surgery from third-party payer and patient perspectives. Critical parameters identified by uncertainty analysis were rates of local and distant recurrence in W&W, salvage surgery, perioperative mortality and utilities assigned to W&W and surgery. CONCLUSION Despite heterogenous methodological design and reporting quality, W&W is likely to be cost effective and cost saving compared with standard care following cCR in LARC. Clinical Trials Registration PROSPERO CRD42024513874.
Collapse
Affiliation(s)
- Ishraq Murshed
- Discipline of Surgery, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.
- Colorectal Unit, Department of Surgery, Royal Adelaide Hospital, Adelaide, SA, Australia.
| | - Zachary Bunjo
- Discipline of Surgery, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Warren Seow
- Discipline of Surgery, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Ishmam Murshed
- Discipline of Surgery, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Sergei Bedrikovetski
- Colorectal Unit, Department of Surgery, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Michelle Thomas
- Discipline of Surgery, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Colorectal Unit, Department of Surgery, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Tarik Sammour
- Discipline of Surgery, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Colorectal Unit, Department of Surgery, Royal Adelaide Hospital, Adelaide, SA, Australia
| |
Collapse
|
7
|
Seven İ, Aktürk Esen S, Sekmek S, Karahan İ, Büyükaksoy M, Kökenek Ünal TD, Uncu D. Clinicopathological features and treatment outcomes of urothelial carcinoma variant histologies and non-urothelial bladder cancers. Int Urol Nephrol 2024:10.1007/s11255-024-04341-w. [PMID: 39729262 DOI: 10.1007/s11255-024-04341-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
PURPOSE Most bladder cancers are pure urothelial carcinomas, but a small portion, approximately 5-10%, have variant histology or are non-urothelial in nature. This research sought to examine the features of and treatment strategies for different types of urothelial carcinoma with variant histologies and non-urothelial bladder cancer. METHODS The study cohort comprised individuals with non-urothelial and variant urothelial bladder cancers treated at two medical centres in Ankara, Turkey, between 2005 and 2024. RESULTS A total of 104 individuals were reviewed, with 88 having urothelial cancer with variant histology and 16 having non-urothelial cancer. Non-urothelial cancers included neuroendocrine, undifferentiated, adenocarcinoma, squamous, sarcoma, and carcinosarcoma, with a median overall survival (OS) of 8 months. The most frequent urothelial carcinoma variants were squamous (43 cases), plasmacytoid (9 cases), and sarcomatoid (6 cases). Individuals with operable variants of urothelial malignancies had a median disease-free survival (DFS) of 16.5 months, while individuals with inoperable/metastatic variants experienced a median progression-free survival (PFS) of 8.9 months. The median OS in the operable cohort was 18.5 months, compared to 10.8 months in the inoperable/metastatic group. CONCLUSION The present study reveals that variant urothelial and non-urothelial bladder cancers are aggressive in nature and have poor prognosis. Given the significant heterogeneity observed in OS, DFS, and PFS among these rare and diverse tumor subtypes, large-scale multicenter investigations are required to establish a consensus on patient handling and treatment.
Collapse
Affiliation(s)
- İsmet Seven
- Medical Oncology Clinic, Ankara Bilkent City Hospital, 06100, Çankaya, Ankara, Turkey.
| | - Selin Aktürk Esen
- Medical Oncology Clinic, Ankara Bilkent City Hospital, 06100, Çankaya, Ankara, Turkey
| | - Serhat Sekmek
- Medical Oncology Clinic, Ankara Bilkent City Hospital, 06100, Çankaya, Ankara, Turkey
| | - İrfan Karahan
- Medical Oncology Clinic, Ankara Bilkent City Hospital, 06100, Çankaya, Ankara, Turkey
| | - Müge Büyükaksoy
- Ankara Bilkent City Hospital, Internal Medicine Clinic, Ankara, Turkey
| | | | - Doğan Uncu
- Medical Oncology Clinic, Ankara Bilkent City Hospital, 06100, Çankaya, Ankara, Turkey
| |
Collapse
|
8
|
Sekmek S, Ozsan Çelebi SN, Bayram D, Erol C, Kos FT, Sendur MAN, Altıntas YE, Tuylu T, Yildirim S, Biter S, Kıdı MM, Bayram E, Majidova N, Bayoglu IV, Atak M, Baskurt K, Akbas S, Alkan A, Bayramgil A, Aslan F, Sahin E, Balcik OY, Bayhan AZ, Saray S, Arpaci E, Ergun Y. Bevacizumab versus aflibercept with FOLFIRI after FOLFOX and bevacizumab in RAS mutant metastatic colon cancer a Turkish oncology group study. Sci Rep 2024; 14:29785. [PMID: 39616250 PMCID: PMC11608356 DOI: 10.1038/s41598-024-81371-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
We aimed to compare FOLFIRI and bevacizumab with FOLFIRI and aflibercept in terms of overall survival (OS), progression-free survival (PFS) and safety in patients with RAS-mutant metastatic colon cancer who progressed after first-line FOLFOX or XELOX and bevacizumab treatment. This retrospective study included 243 patients from 15 different centres in Turkey. The endpoints of the study were OS, PFS and safety and side effect outcomes. The median age of the patients included in the study was 60 (21-85) years. Of the patients enrolled in the study, 114 patients (46.9%) received aflibercept and 129 patients (53.1%) received bevacizumab. Median OS was 11.2 (95% CI: 9.1-13.2) months in patients receiving FOLFIRI + aflibercept and 14.1 (95% CI: 11.2-17.1) months in patients receiving FOLFIRI + bevacizumab. The median PFS was 5.7 (95% CI: 4.9-6.5) months in the aflibercept arm and 7.7 (95% CI: 7.1-8.3) months in the bevacizumab arm. Grade 3-4 side effects were observed in 58 (50.9%) patients in the aflibercept arm and 33 (25.6%) patients in the bevacizumab arm. As a result of our study, in patients with metastatic RAS-mutant colon cancer who progressed after first-line oxaliplatin-based doublet chemotherapy and bevacizumab, better OS and PFS results were obtained in patients receiving bevacizumab with FOLFIRI compared to patients receiving aflibercept with FOLFIRI. In addition, the side effect profile was more tolerable in the bevacizumab arm.
Collapse
Affiliation(s)
- Serhat Sekmek
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey.
| | | | - Dogan Bayram
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey
| | - Cihan Erol
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey
| | - Fahriye Tugba Kos
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey
| | | | - Yunus Emre Altıntas
- Department of Medical Oncology, Kartal Dr. Lutfi Kirdar City Hospital, Istanbul, Turkey
| | - Tugba Tuylu
- Department of Medical Oncology, Kartal Dr. Lutfi Kirdar City Hospital, Istanbul, Turkey
| | - Sedat Yildirim
- Department of Medical Oncology, Kartal Dr. Lutfi Kirdar City Hospital, Istanbul, Turkey
| | - Sedat Biter
- Department of Medical Oncology, Cukurova University, Adana, Turkey
| | | | - Ertugrul Bayram
- Department of Medical Oncology, Cukurova University, Adana, Turkey
| | - Nargiz Majidova
- Department of Medical Oncology, Marmara University, Istanbul, Turkey
| | | | - Mehmetcan Atak
- Department of Medical Oncology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Kadriye Baskurt
- Department of Medical Oncology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Sinem Akbas
- Department of Medical Oncology, Koc University, Istanbul, Turkey
| | - Ali Alkan
- Department of Medical Oncology, Mugla Sıtkı Kocman University Faculty of Medicine, Mugla, Turkey
| | - Ayberk Bayramgil
- Department of Medical Oncology, Umraniye Training and Research Hospital, Istanbul, Turkey
| | - Ferit Aslan
- Department of Medical Oncology, Medicalpark Ankara Batikent Hospital, Ankara, Turkey
| | - Elif Sahin
- Department of Medical Oncology, Kocaeli City Hospital, Kocaeli, Turkey
| | - Onur Yazdan Balcik
- Department of Medical Oncology, Mardin Training and Research Hospital, Mardin, Turkey
| | - Ahmet Ziya Bayhan
- Department of Medical Oncology, Sivas Numune Hospital, Sivas, Turkey
| | - Seray Saray
- Department of Medical Oncology, Balikesir Ataturk City Hospital, Balıkesir, Turkey
| | - Erkan Arpaci
- Department of Medical Oncology, Sakarya Adatip Hospital, Sakarya, Turkey
| | - Yakup Ergun
- Department of Medical Oncology, Bower Hospital, Diyarbakir, Turkey
| |
Collapse
|
9
|
Yousef M, Yousef A, Chowdhury S, Fanaeian MM, Knafl M, Peterson J, Zeineddine M, Alfaro K, Zeineddine F, Goldstein D, Hornstein N, Dasari A, Huey R, Johnson B, Higbie V, Bent A, Kee B, Lee M, Morelli MP, Morris VK, Halperin D, Overman MJ, Parseghian C, Vilar E, Wolff R, Raghav KP, White MG, Uppal A, Sun R, Wang W, Kopetz S, Willis J, Shen JP. Molecular, Socioeconomic, and Clinical Factors Affecting Racial and Ethnic Disparities in Colorectal Cancer Survival. JAMA Oncol 2024; 10:1519-1529. [PMID: 39264607 PMCID: PMC11393757 DOI: 10.1001/jamaoncol.2024.3666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/05/2024] [Indexed: 09/13/2024]
Abstract
Importance Disparity in overall survival (OS) and differences in the frequency of driver gene variants by race and ethnicity have been separately observed in patients with colorectal cancer; however, how these differences contribute to survival disparity is unknown. Objective To quantify the association of molecular, socioeconomic, and clinical covariates with racial and ethnic disparities in overall survival among patients with colorectal cancer. Design, Setting, and Participants This single-center cohort study was conducted at a tertiary-level cancer center using relevant data on all patients diagnosed with colorectal cancer from January 1, 1973, to March 1, 2023. The relative contribution of variables to the disparity was determined using mediation analysis with sequential multivariate Cox regression models. Main Outcome OS, from diagnosis date and from start of first-line chemotherapy. Results The study population of 47 178 patients (median [IQR] age, 57.0 [49-66] years; 20 465 [43.4%] females and 26 713 [56.6%] males; 3.0% Asian, 8.7% Black, 8.8% Hispanic, and 79.4% White individuals) had a median (IQR) follow-up from initial diagnosis of 124 (174) months and OS of 55 (145) months. Compared with White patients, Black patients had worse OS (hazard ratio [HR], 1.16; 95% CI, 1.09-1.24; P <.001), whereas Asian and Hispanic patients had better OS (HR, 0.66; 95% CI, 0.59-0.74; P <.001; and 0.86; 95% CI, 0.81-0.92; P <.001, respectively). When restricted to patients with metastatic disease, the greatest disparity was between Black patients compared with White patients (HR, 1.2; 95% CI, 1.06-1.37; P <.001). Evaluating changes in OS disparity over 20 years showed disparity decreasing among Asian, Hispanic, and White patients, but increasing between Black patients and White patients (HRs, 1.18; 95% CI, 1.07-1.31 for 2008-2012; 1.24, 95% CI, 1.08-1.42 for 2013-2017; and 1.50; 95% CI, 1.20-1.87 for 2018-2023). Survival outcomes for first-line chemotherapy were worse for Black patients compared with White patients (median OS, 18 vs 26 months; HR, 1.30; 95% CI, 1.01-1.70). Among 7628 patients who had clinical molecular testing, APC, KRAS, and PIK3CA showed higher variant frequency in Black patients (false discovery rate [FDR], 0.01; < 0.001; and 0.01, respectively), whereas BRAF and KIT were higher among White patients (FDR, 0.001 and 0.01). Mediation analysis identified neighborhood socioeconomic status as the greatest contributor to OS disparity (29%), followed by molecular characteristics (microsatellite instability status, KRAS variation and BRAF variation, 10%), and tumor sidedness (9%). Conclusions This single-center cohort study identified substantial OS disparity and differing frequencies of driver gene variations by race and ethnicity. Socioeconomic status had the largest contribution but accounted for less than one-third of the disparity, with substantial contribution from tumor molecular features. Further study of the associations of genetic ancestry and the molecular pathogenesis of colorectal cancer with chemotherapy response is needed.
Collapse
Affiliation(s)
- Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Abdelrahman Yousef
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Saikat Chowdhury
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Mohammad M. Fanaeian
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Mark Knafl
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston
| | - Jennifer Peterson
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Mohammad Zeineddine
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Kristin Alfaro
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Fadl Zeineddine
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | | | - Nicholas Hornstein
- Department of General Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Ryan Huey
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Benny Johnson
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Victoria Higbie
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Alisha Bent
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Bryan Kee
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Michael Lee
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Maria Pia Morelli
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Van Karlyle Morris
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Daniel Halperin
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Christine Parseghian
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston
| | - Robert Wolff
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Kanwal P. Raghav
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Michael G. White
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston
| | - Abhineet Uppal
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston
| | - Ryan Sun
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston
| | - Wenyi Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Jason Willis
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
10
|
Anselmino LE, Malizia F, Avila A, Cesatti Laluce N, Mamberto M, Zanotti LC, Farré C, Sauzeau V, Menacho Márquez M. Overcoming Therapy Resistance in Colorectal Cancer: Targeting the Rac1 Signaling Pathway as a Potential Therapeutic Approach. Cells 2024; 13:1776. [PMID: 39513883 PMCID: PMC11545287 DOI: 10.3390/cells13211776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/10/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Colorectal cancer (CRC) is the third most commonly diagnosed type of cancer worldwide and is responsible for numerous deaths. 5-fluorouracil (5-FU) is an effective chemotherapy drug commonly used in the treatment of CRC, either as monotherapy or in combination with other drugs. However, half of CRC cases are resistant to 5-FU-based therapies. To contribute to the understanding of the mechanisms underlying CRC resistance or recurrence after 5-FU-based therapies, we performed a comprehensive study integrating in silico, in vitro, and in vivo approaches. We identified differentially expressed genes and enrichment of pathways associated with recurrence after 5-FU-based therapies. Using these bioinformatics data as a starting point, we selected a group of drugs that restored 5-FU sensitivity to 5-FU resistant cells. Interestingly, treatment with the novel Rac1 inhibitor, 1A-116, reversed morphological changes associated with 5-FU resistance.. Moreover, our in vivo studies have shown that 1A-116 affected tumor growth and the development of metastasis. All our data allowed us to postulate that targeting Rac1 represents a promising avenue for the development of new treatments for patients with CRC resistant to 5-FU-based therapies.
Collapse
Affiliation(s)
- Luciano E. Anselmino
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 2000, Argentina; (L.E.A.); (F.M.); (N.C.L.); (M.M.); (L.C.Z.); (C.F.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 2000, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| | - Florencia Malizia
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 2000, Argentina; (L.E.A.); (F.M.); (N.C.L.); (M.M.); (L.C.Z.); (C.F.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 2000, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| | - Aylén Avila
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| | - Nahuel Cesatti Laluce
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 2000, Argentina; (L.E.A.); (F.M.); (N.C.L.); (M.M.); (L.C.Z.); (C.F.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 2000, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| | - Macarena Mamberto
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 2000, Argentina; (L.E.A.); (F.M.); (N.C.L.); (M.M.); (L.C.Z.); (C.F.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 2000, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| | - Lucía C. Zanotti
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 2000, Argentina; (L.E.A.); (F.M.); (N.C.L.); (M.M.); (L.C.Z.); (C.F.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 2000, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| | - Cecilia Farré
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 2000, Argentina; (L.E.A.); (F.M.); (N.C.L.); (M.M.); (L.C.Z.); (C.F.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 2000, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| | - Vincent Sauzeau
- Institut du Thorax, Inserm, CNRS, Université de Nantes, 44000 Nantes, France;
| | - Mauricio Menacho Márquez
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 2000, Argentina; (L.E.A.); (F.M.); (N.C.L.); (M.M.); (L.C.Z.); (C.F.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 2000, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| |
Collapse
|
11
|
Huang WL, Hsu YC, Luo CW, Chang SJ, Hung YH, Lai CY, Yang YT, Chen YZ, Wu CC, Chen FM, Hou MF, Pan MR. Targeting the CDK7-MDK axis to suppresses irinotecan resistance in colorectal cancer. Life Sci 2024; 353:122914. [PMID: 39004275 DOI: 10.1016/j.lfs.2024.122914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
AIMS Colorectal cancer (CRC) remains a major global health issue, with metastatic cases presenting poor prognosis despite advances in chemotherapy and targeted therapy. Irinotecan, a key drug for advanced CRC treatment, faces challenges owing to the development of resistance. This study aimed to understand the mechanisms underlying irinotecan resistance in colorectal cancer. MAIN METHODS We created a cell line resistant to irinotecan using HT29 cells. These resistant cells were utilized to investigate the role of the CDK7-MDK axis. We employed bulk RNA sequencing, conducted in vivo experiments with mice, and analyzed patient tissues to examine the effects of the CDK7-MDK axis on the cellular response to irinotecan. KEY FINDINGS Our findings revealed that HT29 cells resistant to irinotecan, a crucial colorectal cancer medication, exhibited significant phenotypic and molecular alterations compared to their parental counterparts, including elevated stem cell characteristics and increased levels of cytokines and drug resistance proteins. Notably, CDK7 expression was substantially higher in these resistant cells, and targeting CDK7 effectively decreased their survival and tumor growth, enhancing irinotecan sensitivity. RNA-seq analysis indicated that suppression of CDK7 in irinotecan-resistant HT29 cells significantly reduced Midkine (MDK) expression. Decreased CDK7 and MDK levels, achieved through siRNA and the CDK7 inhibitor THZ1, enhanced the sensitivity of resistant HT29 cells to irinotecan. SIGNIFICANCE Our study sheds light on how CDK7 and MDK influence irinotecan resistance in colorectal and highlights the potential of MDK-targeted therapies. We hypothesized that irinotecan sensitivity and overall treatment efficacy would improve by inhibiting MDK. This finding encourages a careful yet proactive investigation of MDK as a therapeutic target to enhance outcomes in colorectal cancer patients.
Collapse
Affiliation(s)
- Wei-Lun Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Radiation Oncology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
| | - Yin-Chou Hsu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University, Kaohsiung City 824, Taiwan
| | - Chi-Wen Luo
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung City 807, Taiwan; Department of Cosmetic Science, Institute of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan.
| | - Shu-Jyuan Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Hsuan Hung
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Chiao-Ying Lai
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Tzu Yang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yi-Zi Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chun-Chieh Wu
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Fang-Ming Chen
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung City 807, Taiwan
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung City 807, Taiwan; Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Mei-Ren Pan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung City 807, Taiwan.
| |
Collapse
|
12
|
Ota M, Taniguchi K, Hori M, Katanoda K, Nakata K, Miyashiro I, Matsuda T, Lee S, Ito Y. Trends in patterns of treatment and survival of colorectal cancer patients using cancer registry data in Japan: 1995-2015. Cancer Sci 2024; 115:2786-2794. [PMID: 38715379 PMCID: PMC11309936 DOI: 10.1111/cas.16210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 08/10/2024] Open
Abstract
Recent advances in treating colorectal cancer (CRC) have increased the importance of multidisciplinary treatment. This study aimed to clarify trends in the treatment and survival of CRC using population-based cancer registry data in Japan. We analyzed the survival of CRC cases diagnosed from 1995 through 2015 from a population-based cancer registry of six prefectures. The year of diagnosis was classified into five periods, and the trends in the detailed categorization of treatments and survival were identified. We calculated net survival and excess hazard of death from cancer using data on 256,590 CRC patients. The use of laparoscopic surgery has been increasing since 2005 and accounts for the largest proportion of treatment types in the most recent period. Net survival of CRC patients diagnosed after 2005 remained high for laparoscopic surgery and endoscopic surgery (endoscopic mucosal resection or endoscopic submucosal dissection). There was an upward trend in treatment with chemotherapy in addition to open and laparoscopic surgery. Using the excess hazard ratio at the regional stage since 2005, there has been a significant improvement in survival in the younger age group and the rectum cancer group. By type of treatment, there was a tendency toward significant improvement in the open surgery + chemotherapy group. We clarified the trends in treating CRC and the associated trends in survival. Continuous survey based on population-based data helps monitor the impact of developments in treatment.
Collapse
Affiliation(s)
- Masato Ota
- Department of General and Gastroenterological SurgeryOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
- Center for Medical Research & Development, Division of Translational ResearchOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
| | - Kohei Taniguchi
- Translational Research ProgramOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
| | - Megumi Hori
- School of NursingUniversity of ShizuokaShizuoka CityJapan
| | - Kota Katanoda
- National Cancer Center Institute for Cancer ControlTokyoJapan
| | - Kayo Nakata
- Cancer Control Center, Osaka International Cancer InstituteOsakaJapan
| | - Isao Miyashiro
- Cancer Control Center, Osaka International Cancer InstituteOsakaJapan
| | | | - Sang‐Woong Lee
- Department of General and Gastroenterological SurgeryOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
| | - Yuri Ito
- Center for Medical Research & Development, Division of Translational ResearchOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
| |
Collapse
|
13
|
Zhan Y, Cheng X, Mei P, Tan S, Feng W, Jiang H. Safety of first-line systemic therapy in patients with metastatic colorectal cancer: a network meta-analysis of randomized controlled trials. BMC Cancer 2024; 24:893. [PMID: 39048944 PMCID: PMC11270896 DOI: 10.1186/s12885-024-12662-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 07/19/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVE To evaluate the safety of first-line systemic therapy for metastatic colorectal cancer through network meta-analysis. METHODS The literature from PubMed, Embase, Web of Science, and Cochrane Library databases was searched from the inception of the databases to August 15, 2023, and strict inclusion and exclusion criteria were applied to screen studies. The Cochrane Bias Risk Assessment Tool (RoB 2.0) was used to evaluate the quality of the included literature. Network meta-analysis was conducted using Stata 15.0 and R4.3.1 software to compare the incidence of adverse events (AEs) among different treatment regimens. RESULTS A total of 53 randomized controlled trials, involving 17,351 patients with metastatic colorectal cancer (mCRC), were ultimately included, encompassing 29 different therapeutic approaches. According to SUCRA rankings, the CAPOX regimen is most likely to rank first in terms of safety, while the FOLFOXIRI + panitumumab regimen is most likely to rank last. In terms of specific AEs, the CAPOX regimen, whether used alone or in combination with targeted drugs (bevacizumab and cetuximab), is associated with a reduced risk of neutropenia and febrile neutropenia, as well as an increased risk of thrombocytopenia and diarrhea. The FOLFOX regimen, with or without bevacizumab, is linked to an increased risk of neutropenia and peripheral sensory neuropathy. The FOLFIRI/CAPIRI + bevacizumab regimen is associated with a reduced risk of peripheral sensory neuropathy. S-1 and S-1 + oxaliplatin are well-tolerated in terms of gastrointestinal reactions. The FOLFOXIRI regimen, whether used alone or in combination with targeted drugs, is associated with various AEs. CONCLUSION In summary, the CAPOX regimen may be the safest option among the first-line systemic treatment regimens for mCRC patients, while the FOLFOXIRI + panitumumab regimen may be associated with a higher incidence of grade 3 or higher AEs.
Collapse
Affiliation(s)
- Yanrong Zhan
- Rudong People's Hospital / Affiliated Rudong Hospital of Xinglin College, Nantong University, Nantong, Jiangsu, 226400, China.
| | - Xianwen Cheng
- Ankang Hospital of Traditional Chinese Medicine, Ankang, Shaanxi, 725000, China
| | - Pingping Mei
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shufa Tan
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712000, China
| | - Wenzhe Feng
- Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712000, China.
| | - Hua Jiang
- Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712000, China
| |
Collapse
|
14
|
Karlsson JOG, Jynge P. Manganese- and Platinum-Driven Oxidative and Nitrosative Stress in Oxaliplatin-Associated CIPN with Special Reference to Ca 4Mn(DPDP) 5, MnDPDP and DPDP. Int J Mol Sci 2024; 25:4347. [PMID: 38673932 PMCID: PMC11050347 DOI: 10.3390/ijms25084347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Platinum-containing chemotherapeutic drugs are efficacious in many forms of cancer but are dose-restricted by serious side effects, of which peripheral neuropathy induced by oxidative-nitrosative-stress-mediated chain reactions is most disturbing. Recently, hope has been raised regarding the catalytic antioxidants mangafodipir (MnDPDP) and calmangafodipir [Ca4Mn(DPDP)5; PledOx®], which by mimicking mitochondrial manganese superoxide dismutase (MnSOD) may be expected to overcome oxaliplatin-associated chemotherapy-induced peripheral neuropathy (CIPN). Unfortunately, two recent phase III studies (POLAR A and M trials) applying Ca4Mn(DPDP)5 in colorectal cancer (CRC) patients receiving multiple cycles of FOLFOX6 (5-FU + oxaliplatin) failed to demonstrate efficacy. Instead of an anticipated 50% reduction in the incidence of CIPN in patients co-treated with Ca4Mn(DPDP)5, a statistically significant increase of about 50% was seen. The current article deals with confusing differences between early and positive findings with MnDPDP in comparison to the recent findings with Ca4Mn(DPDP)5. The POLAR failure may also reveal important mechanisms behind oxaliplatin-associated CIPN itself. Thus, exacerbated neurotoxicity in patients receiving Ca4Mn(DPDP)5 may be explained by redox interactions between Pt2+ and Mn2+ and subtle oxidative-nitrosative chain reactions. In peripheral sensory nerves, Pt2+ presumably leads to oxidation of the Mn2+ from Ca4Mn(DPDP)5 as well as from Mn2+ in MnSOD and other endogenous sources. Thereafter, Mn3+ may be oxidized by peroxynitrite (ONOO-) into Mn4+, which drives site-specific nitration of tyrosine (Tyr) 34 in the MnSOD enzyme. Conformational changes of MnSOD then lead to the closure of the superoxide (O2•-) access channel. A similar metal-driven nitration of Tyr74 in cytochrome c will cause an irreversible disruption of electron transport. Altogether, these events may uncover important steps in the mechanism behind Pt2+-associated CIPN. There is little doubt that the efficacy of MnDPDP and its therapeutic improved counterpart Ca4Mn(DPDP)5 mainly depends on their MnSOD-mimetic activity when it comes to their potential use as rescue medicines during, e.g., acute myocardial infarction. However, pharmacokinetic considerations suggest that the efficacy of MnDPDP on Pt2+-associated neurotoxicity depends on another action of this drug. Electron paramagnetic resonance (EPR) studies have demonstrated that Pt2+ outcompetes Mn2+ and endogenous Zn2+ in binding to fodipir (DPDP), hence suggesting that the previously reported protective efficacy of MnDPDP against CIPN is a result of chelation and elimination of Pt2+ by DPDP, which in turn suggests that Mn2+ is unnecessary for efficacy when it comes to oxaliplatin-associated CIPN.
Collapse
Affiliation(s)
- Jan Olof G. Karlsson
- Department of Biomedical and Clinical Sciences, Division of Clinical Chemistry and Pharmacology, Linköping University, 581 83 Linköping, Sweden
| | - Per Jynge
- Department of Radiology, Innlandet Trust Hospital, Gjøvik Hospital, 2819 Gjøvik, Norway;
| |
Collapse
|
15
|
Half EE, Levi Z, Mannalithara A, Leshno M, Ben-Aharon I, Abu-Freha N, Silverman B, Ladabaum U. Colorectal cancer screening at age 45 years in Israel: Cost-effectiveness and global implications. Cancer 2024; 130:901-912. [PMID: 38180788 DOI: 10.1002/cncr.35097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) incidence at ages <50 years is increasing worldwide. Screening initiation was lowered to 45 years in the United States. The cost-effectiveness of initiating CRC screening at 45 years in Israel was assessed with the aim of informing national policy and addressing internationally relevant questions. METHODS A validated CRC screening model was calibrated to Israeli data and examined annual fecal immunochemical testing (FIT) or colonoscopy every 10 years from 45 to 74 years (FIT45-74 or Colo45-74) versus from 50 to 74 years (FIT50-74 or Colo50-74). The addition of a fourth colonoscopy at 75 years was explored, subanalyses were performed by sex/ethnicity, and resource demands were estimated. RESULTS FIT50-74 and Colo50-74 reduced CRC incidence by 57% and 70% and mortality by 70% and 77%, respectively, versus no screening, with greater absolute impact in Jews/Other versus Arabs but comparable relative impact. FIT45-74 further reduced CRC incidence and mortality by an absolute 3% and 2%, respectively. With Colo45-74 versus Colo50-74, CRC cases and deaths increased slightly as three colonoscopies per lifetime shifted to 5 years earlier but mean quality-adjusted life-years gained (QALYGs) per person increased. FIT45-74 and Colo45-74 cost 23,800-53,900 new Israeli shekels (NIS)/QALYG and 110,600-162,700 NIS/QALYG, with the lowest and highest values among Jewish/Other men and Arab women, respectively. A fourth lifetime colonoscopy cost 48,700 NIS/QALYG. Lowering FIT initiation to 45 years with modest participation required 19,300 additional colonoscopies in the first 3 years. CONCLUSIONS Beginning CRC screening at 45 years in Israel is projected to yield modest clinical benefits at acceptable costs per QALYG. Despite different estimates by sex/ethnicity, a uniform national policy is favored. These findings can inform Israeli guidelines and serve as a case study internationally.
Collapse
Affiliation(s)
- Elizabeth E Half
- Gastroenterology Institute, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Zohar Levi
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ajitha Mannalithara
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford School of Medicine, Stanford University, Stanford, California, USA
| | - Moshe Leshno
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Coller School of Management, Tel Aviv University, Tel Aviv, Israel
| | - Irit Ben-Aharon
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Division of Oncology, Rambam Health Care Campus, Haifa, Israel
| | - Naim Abu-Freha
- Department of Gastroenterology and Hepatology, Soroka University Medical Center, Beer Sheva, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Barbara Silverman
- Israel National Cancer Registry, Ministry of Health, Ramat Gan, Israel
| | - Uri Ladabaum
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford School of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
16
|
Saadh MJ, Allela OQB, Sattay ZJ, Al Zuhairi RAH, Ahmad H, Eldesoky GE, Adil M, Ali MS. Deciphering the functional landscape and therapeutic implications of noncoding RNAs in the TGF-β signaling pathway in colorectal cancer: A comprehensive review. Pathol Res Pract 2024; 255:155158. [PMID: 38320438 DOI: 10.1016/j.prp.2024.155158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 02/08/2024]
Abstract
Colorectal cancer (CRC) remains a major global health concern, necessitating an in-depth exploration of the intricate molecular mechanisms underlying its progression and potential therapeutic interventions. Transforming Growth Factor-β (TGF-β) signaling, a pivotal pathway implicated in CRC plays a dual role as a tumor suppressor in the early stages and a promoter of tumor progression in later stages. Recent research has shed light on the critical involvement of noncoding RNAs (ncRNAs) in modulating the TGF-β signaling pathway, introducing a new layer of complexity to our understanding of CRC pathogenesis. This comprehensive review synthesizes the current state of knowledge regarding the function and therapeutic potential of various classes of ncRNAs, including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), in the context of TGF-β signaling in CRC. The intricate interplay between these ncRNAs and key components of the TGF-β pathway is dissected, revealing regulatory networks that contribute to the dynamic balance between tumor suppression and promotion. Emphasis is placed on how dysregulation of specific ncRNAs can disrupt this delicate equilibrium, fostering CRC initiation, progression, and metastasis. Moreover, the review provides a critical appraisal of the emerging therapeutic strategies targeting ncRNAs associated with TGF-β signaling in CRC. The potential of these ncRNAs as diagnostic and prognostic biomarkers is discussed, highlighting their clinical relevance. Additionally, the challenges and prospects of developing RNA-based therapeutics, such as RNA interference and CRISPR/Cas-based approaches, are explored in the context of modulating TGF-β signaling for CRC treatment. In conclusion, this review offers a comprehensive overview of the intricate interplay between ncRNAs and the TGF-β signaling pathway in CRC. By unraveling the functional significance of these regulatory elements, we gain valuable insights into the molecular landscape of CRC, paving the way for the development of novel and targeted therapeutic interventions aimed at modulating the TGF-β signaling cascade through the manipulation of ncRNAs.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | | | - Zahraa Jasim Sattay
- Department of Medical Laboratory Technology l, University of imam Jaafar Al-Sadiq, Iraq
| | | | - Hijaz Ahmad
- Section of Mathematics, International Telematic University Uninettuno, Corso Vittorio Emanuele II, 39, Rome 00186, Italy; Center for Applied Mathematics and Bioinformatics, Gulf University for Science and Technology, Kuwait; Department of Computer Science and Mathematics, Lebanese American University, Beirut, Lebanon
| | - Gaber E Eldesoky
- Chemistry Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | | | | |
Collapse
|
17
|
Manickasamy MK, Jayaprakash S, Girisa S, Kumar A, Lam HY, Okina E, Eng H, Alqahtani MS, Abbas M, Sethi G, Kumar AP, Kunnumakkara AB. Delineating the role of nuclear receptors in colorectal cancer, a focused review. Discov Oncol 2024; 15:41. [PMID: 38372868 PMCID: PMC10876515 DOI: 10.1007/s12672-023-00808-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/20/2023] [Indexed: 02/20/2024] Open
Abstract
Colorectal cancer (CRC) stands as one of the most prevalent form of cancer globally, causing a significant number of deaths, surpassing 0.9 million in the year 2020. According to GLOBOCAN 2020, CRC ranks third in incidence and second in mortality in both males and females. Despite extensive studies over the years, there is still a need to establish novel therapeutic targets to enhance the patients' survival rate in CRC. Nuclear receptors (NRs) are ligand-activated transcription factors (TFs) that regulate numerous essential biological processes such as differentiation, development, physiology, reproduction, and cellular metabolism. Dysregulation and anomalous expression of different NRs has led to multiple alterations, such as impaired signaling cascades, mutations, and epigenetic changes, leading to various diseases, including cancer. It has been observed that differential expression of various NRs might lead to the initiation and progression of CRC, and are correlated with poor survival outcomes in CRC patients. Despite numerous studies on the mechanism and role of NRs in this cancer, it remains of significant scientific interest primarily due to the diverse functions that various NRs exhibit in regulating key hallmarks of this cancer. Thus, modulating the expression of NRs with their agonists and antagonists, based on their expression levels, holds an immense prospect in the diagnosis, prognosis, and therapeutical modalities of CRC. In this review, we primarily focus on the role and mechanism of NRs in the pathogenesis of CRC and emphasized the significance of targeting these NRs using a variety of agents, which may represent a novel and effective strategy for the prevention and treatment of this cancer.
Collapse
Affiliation(s)
- Mukesh Kumar Manickasamy
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Sujitha Jayaprakash
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore
| | - Huiyan Eng
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, 61421, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, 61421, Abha, Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore.
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
| |
Collapse
|
18
|
Berg SA, McGregor BA. One Size Fits Some: Approaching Rare Malignancies of the Urinary Tract. Curr Treat Options Oncol 2024; 25:206-219. [PMID: 38315403 DOI: 10.1007/s11864-024-01187-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
OPINION STATEMENT Urothelial carcinoma is the predominant cancer of the urinary tract but when divergent and subtype histology (non-urothelial) are identified at time of pathologic diagnosis, therapeutic and diagnostic challenges transpire. To this end, pathologic review to confirm any non-urothelial histology is key since these subtypes can often be overlooked. Few prospective trials are dedicated to understanding these non-urothelial histologic types; however, current, and past trials did allow patients with these non-urothelial histologic types to enroll, and inferences can be made about treatment efficacy and survival. Existing treatment regimens for non-urothelial bladder cancers are akin to standard urothelial cancer regimens using surgical approaches for localized disease and platinum-based chemotherapy for advanced disease. The reported clinical trials, that will be discussed, center on non-urothelial histologic types. These studies, albeit limited, provide critical insight into tumor biology and response to standard platinum-based chemotherapy, immune checkpoint inhibitors, and antibody drug conjugates. The inclusion of non-urothelial histologic types will be essential for clinical trials in development to provide further therapeutic advances and provide essential efficacy data.
Collapse
Affiliation(s)
- Stephanie A Berg
- Dana-Farber Cancer Institute, Lank Center for Genitourinary Oncology, 44 Binney Street, Boston, MA, 02115, USA
| | - Bradley A McGregor
- Dana-Farber Cancer Institute, Lank Center for Genitourinary Oncology, 44 Binney Street, Boston, MA, 02115, USA.
| |
Collapse
|
19
|
Jones VT, Graves-Deal R, Cao Z, Bogatcheva G, Ramirez MA, Harmych SJ, Higginbotham JN, Sharma V, Damalanka VC, Wahoski CC, Joshi N, Irudayam MJ, Roland JT, Ayers GD, Liu Q, Coffey RJ, Janetka JW, Singh B. Inhibition of autocrine HGF maturation overcomes cetuximab resistance in colorectal cancer. Cell Mol Life Sci 2024; 81:28. [PMID: 38212428 PMCID: PMC10784391 DOI: 10.1007/s00018-023-05071-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/27/2023] [Accepted: 11/27/2023] [Indexed: 01/13/2024]
Abstract
Although amplifications and mutations in receptor tyrosine kinases (RTKs) act as bona fide oncogenes, in most cancers, RTKs maintain moderate expression and remain wild-type. Consequently, cognate ligands control many facets of tumorigenesis, including resistance to anti-RTK therapies. Herein, we show that the ligands for the RTKs MET and RON, HGF and HGFL, respectively, are synthesized as inactive precursors that are activated by cellular proteases. Our newly generated HGF/HGFL protease inhibitors could overcome both de novo and acquired cetuximab resistance in colorectal cancer (CRC). Conversely, HGF overexpression was necessary and sufficient to induce cetuximab resistance and loss of polarity. Moreover, HGF-induced cetuximab resistance could be overcome by the downstream MET inhibitor, crizotinib, and upstream protease inhibitors. Additionally, HAI-1, an endogenous inhibitor of HGF proteases, (i) was downregulated in CRC, (ii) exhibited increased genomic methylation that correlated with poor prognosis, (iii) HAI-1 expression correlated with cetuximab response in a panel of cancer cell lines, and (iv) exogenous addition of recombinant HAI-1 overcame cetuximab resistance in CC-HGF cells. Thus, we describe a targetable, autocrine HAI-1/Protease/HGF/MET axis in cetuximab resistance in CRC.
Collapse
Affiliation(s)
- Vivian Truong Jones
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ramona Graves-Deal
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
| | - Zheng Cao
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
| | - Galina Bogatcheva
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
| | - Marisol A Ramirez
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Sarah J Harmych
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - James N Higginbotham
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
| | - Vineeta Sharma
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
| | - Vishnu C Damalanka
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Claudia C Wahoski
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Neeraj Joshi
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
| | - Maria Johnson Irudayam
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
| | - Joseph T Roland
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Gregory D Ayers
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Qi Liu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Robert J Coffey
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - James W Janetka
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Bhuminder Singh
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA.
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
20
|
Qiu L, Chen S, Ben S, Cui J, Lu S, Qu R, Lv J, Shao W, Yu Q. Genetic variants in primary cilia-related genes associated with the prognosis of first-line chemotherapy in colorectal cancer. Cancer Med 2024; 13:e6996. [PMID: 38334481 PMCID: PMC10854446 DOI: 10.1002/cam4.6996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/10/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Primary cilia are antenna-like organelles that conduct physical and chemical signals, which affect cell proliferation, migration, and differentiation. Some researchers have reported the correlation between primary cilia-related genes and prognosis of colorectal cancer (CRC). METHODS The association between single nucleotide polymorphisms (SNPs) of primary cilia-related genes and outcome after the first-line chemotherapy was explored by the Cox regression model. Expression qualitative trait locus (eQTL) analysis was performed to explore the impact of SNPs on gene expression. Tumor Immune Estimation Resource and TISIDB databases were used for investigating the relevance between ODF2L and tumor infiltration immune cells and immunomodulators. RESULTS We identified that rs4288473 C allele of ODF2L had poor progression-free survival (PFS) and overall survival (OS) of CRC patients in the additive model (adjusted HRPFS = 1.39, 95% CI = 1.14-1.70, p = 1.36 × 10-3 , and adjusted HROS = 1.31, 95% CI = 1.03-1.65, p = 2.62 × 10-2 ). The stratified analysis indicated that rs4288573 CC/CT genotype was involved with poor prognosis in the irinotecan-treated subgroup (PPFS = 1.03 × 10-2 , POS = 3.29 × 10-3 ). Besides, ODF2L mRNA expression level was notably up-regrated in CRC tissues. The C allele of rs4288573 was notably related to higher ODF2L mRNA expression levels based on eQTL analysis. Functionally, knockdown of ODF2L inhibited cell proliferation and decrease the chemoresistance of HCT-116 and DLD-1 cells to irinotecan. CONCLUSION Our study indicates that rs4288573 in ODF2L is a potential predictor of the chemotherapy prognosis of CRC.
Collapse
Affiliation(s)
- Lei Qiu
- Department of GastroenterologyThe Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical UniversityNanjingJiangsuChina
| | - Silu Chen
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized MedicineNanjing Medical UniversityNanjingChina
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public HealthNanjing Medical UniversityNanjingChina
| | - Shuai Ben
- Department of Ophthalmology, Shanghai General Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jinxin Cui
- Department of GastroenterologyThe Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical UniversityNanjingJiangsuChina
| | - Shan Lu
- Department of GastroenterologyThe Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical UniversityNanjingJiangsuChina
| | - Rong Qu
- Department of GastroenterologyThe Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical UniversityNanjingJiangsuChina
| | - Jinghuan Lv
- Department of PathologyThe Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical UniversitySuzhouChina
| | - Wei Shao
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized MedicineNanjing Medical UniversityNanjingChina
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public HealthNanjing Medical UniversityNanjingChina
| | - Qiang Yu
- Department of GastroenterologyThe Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical UniversityNanjingJiangsuChina
| |
Collapse
|
21
|
Park HA, Edelmann D, Canzian F, Seibold P, Harrison TA, Hua X, Shi Q, Silverman A, Benner A, Macauda A, Schneider M, Goldberg RM, Alberts SR, Hoffmeister M, Brenner H, Chan AT, Peters U, Newcomb PA, Chang-Claude J. Genome-wide study of genetic polymorphisms predictive for outcome from first-line oxaliplatin-based chemotherapy in colorectal cancer patients. Int J Cancer 2023; 153:1623-1634. [PMID: 37539667 PMCID: PMC10550047 DOI: 10.1002/ijc.34663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 08/05/2023]
Abstract
We conducted the first large genome-wide association study to identify novel genetic variants that predict better (or poorer) prognosis in colorectal cancer patients receiving standard first-line oxaliplatin-based chemotherapy vs chemotherapy without oxaliplatin. We used data from two phase III trials, NCCTG N0147 and NCCTG N9741 and a population-based patient cohort, DACHS. Multivariable Cox proportional hazards models were employed, including an interaction term between each SNP and type of treatment for overall survival (OS) and progression-free survival. The analysis was performed for studies individually, and the results were combined using fixed-effect meta-analyses separately for resected stage III colon cancer (3098 patients from NCCTG N0147 and 549 patients from DACHS) and mCRC (505 patients from NCCTG N9741 and 437 patients from DACHS). We further performed gene-based analysis as well as in silico bioinformatics analysis for CRC-relevant functional genomic annotation of identified loci. In stage III colon cancer patients, a locus on chr22 (rs11912167) was associated with significantly poorer OS after oxaliplatin-based chemotherapy vs chemotherapy without oxaliplatin (Pinteraction < 5 × 10-8 ). For mCRC patients, three loci on chr1 (rs1234556), chr12 (rs11052270) and chr15 (rs11858406) were found to be associated with differential OS (P < 5 × 10-7 ). The locus on chr1 located in the intronic region of RCSD1 was replicated in an independent cohort of 586 mCRC patients from ALGB/SWOG 80405 (Pinteraction = .04). The GWA gene-based analysis yielded for RCSD1 the most significant association with differential OS in mCRC (P = 6.6 × 10-6 ). With further investigation into its biological mechanisms, this finding could potentially be used to individualize first-line treatment and improve clinical outcomes.
Collapse
Affiliation(s)
- Hanla A. Park
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Dominic Edelmann
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Petra Seibold
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tabitha A. Harrison
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United State of America
| | - Xinwei Hua
- Department of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United State of America
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, Massachusetts, United State of America
- Department of Cardiology, Peking University Third Hospital, Peking University, Beijing, China
| | - Qian Shi
- Department of Quantitative Science, Mayo Clinic, Rochester, Minnesota, United State of America
| | - Allison Silverman
- Epidemiology Program, Fred Hutchinson Research Cancer Research Center, Seattle, Washington, United State of America
| | - Axel Benner
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Angelica Macauda
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | | | - Steven R. Alberts
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, United State of America
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andrew T. Chan
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United State of America
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United State of America
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United State of America
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United State of America
- School of Public Health, University of Washington, Seattle, Washington, United State of America
| | - Polly A. Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United State of America
- School of Public Health, University of Washington, Seattle, Washington, United State of America
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Cancer Epidemiology Group, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
22
|
Zhang X, Li F, Li R, Zhao N, Liu D, Xu Y, Wang L, Wang D, Zhao R. B7 Induces Apoptosis in Colorectal Cancer Cells by Regulating the Expression of Caspase-3 and Inhibits Autophagy. Onco Targets Ther 2023; 16:867-883. [PMID: 37915320 PMCID: PMC10617530 DOI: 10.2147/ott.s429128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/18/2023] [Indexed: 11/03/2023] Open
Abstract
Purpose Heterocyclic compounds are organic compounds with heterocyclic structures, which are common in drug molecules. They include pyrazines with diverse functions, including anti-cancer, antimicrobial, antidiabetic, and anticholinergic activities. In this study a new small molecular compound B7 based on tetrazolium substituted pyrazine was synthesized and its effect on the progression of colorectal cancer (CRC) and its potential mechanism were investigated. Methods We synthesized a series of tetrazolium-substituted pyrazine compounds by chemoenzymatic method. NCM460 (Human), HCT116 (Human), SW480 (Human) cell lines were selected to analyse the inhibitory effect of B7 on CRC by CCK-8, apoptosis, cell migration and invasion, qPCR, Western blotting, molecular docking, immunofluorescence. Moreover, a CRC xenograft model of mice was used to analyzed the role of B7 in vivo. Results Among these compounds, 3-methyl-5je-6-bis (1H-tetrazole-5-yl) pyrazine-2-carboxylic acid (B7) inhibited CRC cell proliferation and induced apoptosis. The expression of Caspase-3 was increased after B7 treatment. In addition, the mitochondria abnormalities was observed in B7 group due to decrease the expression of Beclin-1. In addition, B7 inhibited the migration and invasion in CRC cells. Finally, the results showed that B7 had anti-tumor activity in CRC xenograft model of mice. Conclusion In summary, compound B7 was synthesized efficiently using tetrazolium-substituted pyrazine via a chemoenzymatic method. Moreover, B7 have ability to regulate the expression of Caspase-3 which induced apoptosis in CRC cells. In addition, decreased Beclin-1 expression after B7 treatment, indicating inhibited autophagy. This study showed that B7 effectively induced apoptosis and inhibited autophagy in CRC cells.
Collapse
Affiliation(s)
- Xinyi Zhang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, 130062, People’s Republic of China
| | - Fengxi Li
- Key Laboratory of Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130023, People’s Republic of China
| | - Rong Li
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, 130062, People’s Republic of China
| | - Nan Zhao
- Key Laboratory of Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130023, People’s Republic of China
| | - Dianfeng Liu
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, 130062, People’s Republic of China
| | - Yuelin Xu
- Key Laboratory of Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130023, People’s Republic of China
| | - Lei Wang
- Key Laboratory of Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130023, People’s Republic of China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, 130062, People’s Republic of China
| | - Ruihong Zhao
- Department of Gastroenterology Endoscopy Center, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| |
Collapse
|
23
|
van Eerden RAG, de Boer NL, van Kooten JP, Bakkers C, Dietz MV, Creemers GJM, Buijs SM, Bax R, de Man FM, Lurvink RJ, Diepeveen M, Brandt-Kerkhof ARM, van Meerten E, Koolen SLW, de Hingh IHJT, Verhoef C, Mathijssen RHJ, Burger JWA. Phase I study of intraperitoneal irinotecan combined with palliative systemic chemotherapy in patients with colorectal peritoneal metastases. Br J Surg 2023; 110:1502-1510. [PMID: 37467389 DOI: 10.1093/bjs/znad228] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/18/2023] [Accepted: 06/28/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND Patients with colorectal peritoneal metastases who are not eligible for cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) owing to extensive peritoneal disease have a poor prognosis. It was hypothesized that these patients may benefit from the addition of intraperitoneal irinotecan to standard palliative systemic chemotherapy. METHODS This was a classical 3 + 3 phase I dose-escalation trial in patients with colorectal peritoneal metastases who were not eligible for CRS-HIPEC. Intraperitoneal irinotecan was administered every 2 weeks, concomitantly with systemic FOLFOX (5-fluorouracil, folinic acid, oxaliplatin)-bevacizumab. The primary objective was to determine the maximum tolerated dose and dose-limiting toxicities. Secondary objectives were to elucidate the systemic and intraperitoneal pharmacokinetics, safety profile, and efficacy. RESULTS Eighteen patients were treated. No dose-limiting toxicities were observed with 50 mg (4 patients) and 75 mg (9 patients) intraperitoneal irinotecan. Two dose-limiting toxicities occurred with 100 mg irinotecan among five patients. The maximum tolerated dose of intraperitoneal irinotecan was established to be 75 mg, and it was well tolerated. Intraperitoneal exposure to SN-38 (active metabolite of irinotecan) was high compared with systemic exposure (median intraperitoneal area under the curve (AUC) to systemic AUC ratio 4.6). Thirteen patients had a partial radiological response and five had stable disease. Four patients showed a complete response during post-treatment diagnostic laparoscopy. Five patients underwent salvage resection or CRS-HIPEC. Median overall survival was 23.9 months. CONCLUSION Administration of 75 mg intraperitoneal irinotecan concomitantly with systemic FOLFOX-bevacizumab was safe and well tolerated. Intraperitoneal SN-38 exposure was high and prolonged. As oncological outcomes were promising, intraperitoneal administration of irinotecan may be a good alternative to other, more invasive and costly treatment options. A phase II study is currently accruing.
Collapse
Affiliation(s)
- Ruben A G van Eerden
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Nadine L de Boer
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Job P van Kooten
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Checca Bakkers
- Department of Surgery, Catharina Cancer Institute, Eindhoven, the Netherlands
| | - Michelle V Dietz
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Geert-Jan M Creemers
- Department of Medical Oncology, Catharina Cancer Institute, Eindhoven, the Netherlands
| | - Sanne M Buijs
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Ramon Bax
- Department of Medical Oncology, Catharina Cancer Institute, Eindhoven, the Netherlands
| | - Femke M de Man
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Robin J Lurvink
- Department of Surgery, Catharina Cancer Institute, Eindhoven, the Netherlands
| | - Marjolein Diepeveen
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Esther van Meerten
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Stijn L W Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
- Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands
| | | | - Cornelis Verhoef
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Jacobus W A Burger
- Department of Surgery, Catharina Cancer Institute, Eindhoven, the Netherlands
| |
Collapse
|
24
|
Wei X, Chow HY, Chong HC, Leung SL, Ho MK, Lee MY, Leung YC. Arginine Is a Novel Drug Target for Arginine Decarboxylase in Human Colorectal Cancer Cells. Int J Mol Sci 2023; 24:13741. [PMID: 37762044 PMCID: PMC10531272 DOI: 10.3390/ijms241813741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Colorectal cancer (CRC) has been proven to be highly reliant on arginine availability. Limiting arginine-rich foods or treating patients with arginine-depleting enzymes arginine deiminase (ADI) or arginase can suppress colon cancer. However, arginase and ADI are not the best drug candidates for CRC. Ornithine, the product of arginase, can enhance the supply of polyamine, which favors CRC cell growth, while citrulline, the product of ADI, faces the problem of arginine recycling due to the overexpression of argininosuccinate synthetase (ASS). Biosynthetic arginine decarboxylase (ADC), an enzyme that catalyzes the conversion of arginine to agmatine and carbon dioxide, may be a better choice as it combines both arginine depletion and suppression of intracellular polyamine synthesis via its product agmatine. ADC has anti-tumor potential yet has received much less attention than the other two arginine-depleting enzymes. In order to gain a better understanding of ADC, the preparation and the anti-cancer properties of this enzyme were explored in this study. When tested in vitro, ADC inhibited the proliferation of three colorectal cancer cell lines regardless of their ASS cellular expression. In contrast, ADC had a lesser cytotoxic effect on the human foreskin fibroblasts and rat primary hepatocytes. Further in vitro studies revealed that ADC induced S and G2/M phase cell-cycle arrest and apoptosis in HCT116 and LoVo cells. ADC-induced apoptosis in HCT116 cells followed the mitochondrial apoptotic pathway and was caspase-3-dependent. With all results obtained, we suggest that arginine is a potential target for treating colorectal cancer with ADC, and the anti-cancer properties of ADC should be more deeply investigated in the future.
Collapse
Affiliation(s)
- Xinlei Wei
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Ho-Yin Chow
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Hiu-Chi Chong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Siu-Lun Leung
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Mei-Ki Ho
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Man-Yuen Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- Lo Ka Chung Research Centre for Natural Anti-Cancer Drug Development, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Yun-Chung Leung
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- Lo Ka Chung Research Centre for Natural Anti-Cancer Drug Development, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| |
Collapse
|
25
|
Alkader MS, Shahin AA, Alsoreeky MS, Matarweh HB, Abdullah IA. The Impact of Palliative Chemotherapy on the Survival of Patients With Metastatic Colorectal Cancer in Jordan. Cureus 2023; 15:e46187. [PMID: 37790030 PMCID: PMC10544268 DOI: 10.7759/cureus.46187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2023] [Indexed: 10/05/2023] Open
Abstract
Background In Jordan, managing metastatic colorectal cancer (mCRC) is particularly complex, considering limited resources, access to advanced therapies, and unique patient demographics. Palliative chemotherapy, an approach aimed at relieving symptoms and improving the quality of life in patients with advanced cancer, including mCRC, has gained attention as a treatment strategy. While palliative chemotherapy may not aim for complete cancer eradication, it can extend survival, manage disease-related symptoms, and enhance the patient's overall well-being. However, deciding to pursue palliative chemotherapy for mCRC patients involves individual patient characteristics, performance status, disease aggressiveness, potential treatment-related adverse effects, and available healthcare resources. Given the need for region-specific insights into treatment outcomes, the proposed study seeks to investigate the impact of palliative chemotherapy on overall survival (OS), specifically within Jordan's healthcare landscape. Our study aims to showcase palliative chemotherapy's effectiveness on OS in first-line settings. Materials and methods This study is a retrospective analysis conducted at the Military Cancer Center (MCAC) in Jordan. It includes 73 patients diagnosed with mCRC between January 1, 2018, and January 1, 2020. Data were obtained from electronic medical records, and patients were monitored until June 10, 2023. Various patient characteristics were analyzed, including age, sex, primary tumor site, metastatic site, and treatment options for mCRC. The study evaluated the effectiveness of palliative chemotherapy in improving survival rates compared to BSC. Result We conducted a study with 73 participants, whose mean age was 60.37 ±13.5 years and a median of 63. Of these patients, 51 (69.9%) were male, and 22 (30.1%) were female. The primary site of the tumor was located on the left side in 32 patients (43.9%), on the right side in 26 patients (35.6%), and rectal cancer in 15 patients (20.5%). The most common site of the tumor was the sigmoid (17 patients, 23.3%). The liver was the most common site of metastasis (52 patients, 71.2%). Of the patients, 47 (64.4%) received palliative chemotherapy, while 26 (35.6%) were kept on best supportive care (BSC). Of those who received chemotherapy, FOLFIRI was administered to 32 patients (43.8%) and FOLFOX to 15 patients (20.5%). Based on the Kaplan-Meier curve, palliative chemotherapy patients had a significantly longer OS than those who only received BSC. Patients with palliative chemotherapy had a median OS of 12.4 months, while those who only had BSC survived for 5.3 months. The HR was 0.36 with a 95% confidence interval of 0.2-0.62, and the P-value was less than 0.001. Conclusion This study shows that palliative chemotherapy offers a notable advantage and a significant survival benefit compared to BSC.
Collapse
Affiliation(s)
- Mohammad S Alkader
- Department of Medical Oncology, Jordanian Royal Medical Services, Amman, JOR
| | - Ahmed A Shahin
- Department of Medical Oncology, Jordanian Royal Medical Services, Amman, JOR
| | | | - Hanna B Matarweh
- Department of Internal Medicine, Arab Medical Center, Amman, JOR
| | - Ilham A Abdullah
- Department of Internal Medicine, Jordanian Royal Medical Services, Amman, JOR
| |
Collapse
|
26
|
Huang Z, Kaller M, Hermeking H. CRISPR/Cas9-mediated inactivation of miR-34a and miR-34b/c in HCT116 colorectal cancer cells: comprehensive characterization after exposure to 5-FU reveals EMT and autophagy as key processes regulated by miR-34. Cell Death Differ 2023; 30:2017-2034. [PMID: 37488217 PMCID: PMC10406948 DOI: 10.1038/s41418-023-01193-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/26/2023] Open
Abstract
The miR-34a and miR-34b/c encoding genes represent direct targets of the p53 transcription factor, and presumably mediate part of the tumor suppressive effects of p53. Here, we sought to determine their functional relevance by inactivating miR-34a and/or miR-34b/c using a CRISPR/Cas9 approach in the colorectal cancer (CRC) cell line HCT116. Concomitant deletion of miR-34a and miR-34b/c resulted in significantly reduced suppression of proliferation after p53 activation, enhanced migration, invasion and EMT, as well as reduced sensitivity to chemotherapeutics, increased stress-induced autophagic flux, decreased apoptosis and upregulation of autophagy-related genes after 5-FU treatment. However, inactivation of singular miR-34a or miR-34b/c had little effects on the aforementioned processes. RNA-Seq analysis revealed that concomitant deletion of miR-34a/b/c caused EMT signature enrichment, impaired gene repression by the p53-DREAM pathway and elevated autophagy after 5-FU treatment. A gene signature comprised of mRNAs significantly upregulated after combined inactivation of miR-34a and miR-34b/c showed a significant association with the invasive colon cancer subtype CMS4 and poor overall survival in two CRC patient cohorts, and with 5-FU resistance in CRC cell lines. In miR-34a/b/c-deficient cells the upregulated miR-34 target FOXM1 directly induced p62 and ATG9A, which increased autophagy and consequently attenuated apoptosis and rendered the miR-34a/b/c-KO cells more resistant to 5-FU. Inhibition of autophagy by depletion of ATG9A or chloroquine re-sensitized miR-34a/b/c-deficient HCT116 cells to 5-FU. In summary, our findings show a complementary role of miR-34a and miR-34b/c in the regulation of EMT and autophagy which may be relevant for CRC therapy in the future.
Collapse
Affiliation(s)
- Zekai Huang
- Experimental and Molecular Pathology, Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, D-80337, Munich, Germany
| | - Markus Kaller
- Experimental and Molecular Pathology, Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, D-80337, Munich, Germany
| | - Heiko Hermeking
- Experimental and Molecular Pathology, Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, D-80337, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, D-80336, Munich, Germany.
- German Cancer Research Center (DKFZ), D-69120, Heidelberg, Germany.
| |
Collapse
|
27
|
Schrag D, Shi Q, Weiser MR, Gollub MJ, Saltz LB, Musher BL, Goldberg J, Al Baghdadi T, Goodman KA, McWilliams RR, Farma JM, George TJ, Kennecke HF, Shergill A, Montemurro M, Nelson GD, Colgrove B, Gordon V, Venook AP, O'Reilly EM, Meyerhardt JA, Dueck AC, Basch E, Chang GJ, Mamon HJ. Preoperative Treatment of Locally Advanced Rectal Cancer. N Engl J Med 2023; 389:322-334. [PMID: 37272534 PMCID: PMC10775881 DOI: 10.1056/nejmoa2303269] [Citation(s) in RCA: 194] [Impact Index Per Article: 97.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
BACKGROUND Pelvic radiation plus sensitizing chemotherapy with a fluoropyrimidine (chemoradiotherapy) before surgery is standard care for locally advanced rectal cancer in North America. Whether neoadjuvant chemotherapy with fluorouracil, leucovorin, and oxaliplatin (FOLFOX) can be used in lieu of chemoradiotherapy is uncertain. METHODS We conducted a multicenter, unblinded, noninferiority, randomized trial of neoadjuvant FOLFOX (with chemoradiotherapy given only if the primary tumor decreased in size by <20% or if FOLFOX was discontinued because of side effects) as compared with chemoradiotherapy. Adults with rectal cancer that had been clinically staged as T2 node-positive, T3 node-negative, or T3 node-positive who were candidates for sphincter-sparing surgery were eligible to participate. The primary end point was disease-free survival. Noninferiority would be claimed if the upper limit of the two-sided 90.2% confidence interval of the hazard ratio for disease recurrence or death did not exceed 1.29. Secondary end points included overall survival, local recurrence (in a time-to-event analysis), complete pathological resection, complete response, and toxic effects. RESULTS From June 2012 through December 2018, a total of 1194 patients underwent randomization and 1128 started treatment; among those who started treatment, 585 were in the FOLFOX group and 543 in the chemoradiotherapy group. At a median follow-up of 58 months, FOLFOX was noninferior to chemoradiotherapy for disease-free survival (hazard ratio for disease recurrence or death, 0.92; 90.2% confidence interval [CI], 0.74 to 1.14; P = 0.005 for noninferiority). Five-year disease-free survival was 80.8% (95% CI, 77.9 to 83.7) in the FOLFOX group and 78.6% (95% CI, 75.4 to 81.8) in the chemoradiotherapy group. The groups were similar with respect to overall survival (hazard ratio for death, 1.04; 95% CI, 0.74 to 1.44) and local recurrence (hazard ratio, 1.18; 95% CI, 0.44 to 3.16). In the FOLFOX group, 53 patients (9.1%) received preoperative chemoradiotherapy and 8 (1.4%) received postoperative chemoradiotherapy. CONCLUSIONS In patients with locally advanced rectal cancer who were eligible for sphincter-sparing surgery, preoperative FOLFOX was noninferior to preoperative chemoradiotherapy with respect to disease-free survival. (Funded by the National Cancer Institute; PROSPECT ClinicalTrials.gov number, NCT01515787.).
Collapse
Affiliation(s)
- Deborah Schrag
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Qian Shi
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Martin R Weiser
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Marc J Gollub
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Leonard B Saltz
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Benjamin L Musher
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Joel Goldberg
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Tareq Al Baghdadi
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Karyn A Goodman
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Robert R McWilliams
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Jeffrey M Farma
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Thomas J George
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Hagen F Kennecke
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Ardaman Shergill
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Michael Montemurro
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Garth D Nelson
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Brian Colgrove
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Vallerie Gordon
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Alan P Venook
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Eileen M O'Reilly
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Jeffrey A Meyerhardt
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Amylou C Dueck
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Ethan Basch
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - George J Chang
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| | - Harvey J Mamon
- From the Departments of Medicine (D.S., L.B.S., E.M.O.), Surgery (M.R.W.), and Radiology (M.J.G.), Memorial Sloan Kettering Cancer Center, and the Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai (K.A.G.) - both in New York; Alliance Statistics and Data Management Center (Q.S., G.D.N., B.C., A.C.D.) and the Department of Oncology (R.R.M.), Mayo Clinic, Rochester, MN; SWOG Cancer Research Network and the Department of Medicine, Baylor College of Medicine (B.L.M.), and the Department of Colon and Rectal Surgery, M.D. Anderson Cancer Center (G.J.C.) - both in Houston; the Departments of Surgery (J.G.) and Radiation Oncology (H.J.M.), Brigham and Women's Hospital, and the Department of Medical Oncology, Dana-Farber Cancer Institute (J.A.M.) - both in Boston; IHA Hematology Oncology, Ypsilanti, MI (T.A.B.); ECOG-ACRIN Cancer Research Network and Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia (J.M.F.); NRG Oncology and the University of Florida Health Cancer Center, Gainesville (T.J.G.); Canadian Cancer Trials Group, Kingston, ON (H.F.K.), and the Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg (V.G.) - both in Canada; Alliance Protocol Office, Chicago (A.S.); the Swiss Group for Clinical Cancer Research, Bern, Switzerland (M.M.); Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco (A.P.V.); and the Department of Medical Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill (E.B.)
| |
Collapse
|
28
|
Wan C, Ma Q, Anderson S, Zhang QH, Zhang CF, Wang AH, Bell E, Hou L, Yuan CS, Wang CZ. Effects of Curcuminoids and Surfactant-Formulated Curcumin on Chemo-Resistant Colorectal Cancer. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2023; 51:1577-1594. [PMID: 37465963 DOI: 10.1142/s0192415x23500714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related death in the United States, and chronic gut inflammation is a risk factor for CRC initiation and development. Curcuma longa L., or turmeric, has become one of the most studied herbal medicines in recent years due to its anticancer potentials. It is generally accepted that the major component in turmeric is curcuminoids, and the active constituent in curcuminoids is curcumin. However, unprocessed curcumin is characterized by poor water solubility, which means low bioavailability in humans. To increase the bioavailability of curcumin, in this study, we utilized a novel surfactant-formulated curcumin (CuminUP60[Formula: see text]) and evaluated its CRC chemopreventive activities. Compared with the chemo-sensitive CRC cell line HCT-116, the management of the CRC SW-480 cell line is a challenge, since the latter is chemo-resistant. In other words, these cancer cells resist the effects of the chemotherapy. Using the newly formulated CuminUP60[Formula: see text] water solution, this study demonstrated its strong antiproliferative effects on the SW-480 cells in a dose- and time-dependent manner. This new formulation induced early apoptosis and arrested the cell cycle in the G2/M phase via the upregulation of cyclin B1. We also observed that this new formulation possessed inhibitory effects on Th17 cell differentiation, which regulates the body's immune response against gut malignancies. In summary, our results exhibited a potential clinical utility of the surfactant-formulated curcumin in chemo-resistant colorectal cancer management.
Collapse
Affiliation(s)
- Chunping Wan
- Central Laboratory, No. 1, Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming 650021, P. R. China
- Tang Center for Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Qinge Ma
- Tang Center for Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
- Key Laboratory of Modern Preparation of TCM of Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P. R. China
| | - Samantha Anderson
- Tang Center for Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Qi-Hui Zhang
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400044, P. R. China
| | - Chun-Feng Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, P. R. China
| | - Angela H Wang
- Tang Center for Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Emma Bell
- Tang Center for Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Lifei Hou
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Chun-Su Yuan
- Tang Center for Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Chong-Zhi Wang
- Tang Center for Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
29
|
Venook AP. Flashback Foreword: IFL/FOLFOX/IROX in Advanced Colorectal Cancer and FOLFIRI and FOLFOX6 in Colorectal Cancer. J Clin Oncol 2023; 41:3459-3460. [PMID: 37379690 DOI: 10.1200/jco.23.00324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/17/2023] [Indexed: 06/30/2023] Open
Affiliation(s)
- Alan P Venook
- Former Associate Editor, Journal of Clinical Oncology, Alexandria, VA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
30
|
Deshmukh R, Prajapati M, Harwansh RK. A review on emerging targeted therapies for the management of metastatic colorectal cancers. Med Oncol 2023; 40:159. [PMID: 37097307 DOI: 10.1007/s12032-023-02020-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/08/2023] [Indexed: 04/26/2023]
Abstract
Colorectal cancers are among the most commonly found cancers over the world. In spite of the recent advancements in diagnosis and prognosis, the management of this metastatic condition remains a challenge. The utility of monoclonal antibodies in the healing of patients with colorectal cancer has opened a new chapter in the quest for newer therapies. The resistance to the standard treatment regimen made it mandatory to search for newer targets. Mutagenic alterations in the gene engaged in cellular differentiation and growth pathway have been the reason for resistance to treatment. The newer therapies target the various proteins and receptors involved in the signal transduction and down streaming pathways leading to cell proliferation. This review presents an insight into the newer targeted therapies for colorectal cancer involving tyrosine kinase blockers, epidermal growth factor receptors, vascular endothelial growth factor, immune checkpoint therapy, and BRAF inhibitors.
Collapse
Affiliation(s)
- Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India.
| | - Mahendra Prajapati
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| | - Ranjit K Harwansh
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| |
Collapse
|
31
|
Zheng Z, Wu M, Li H, Xu W, Yang M, Pan K, Ni Y, Jiang T, Zheng H, Jin X, Zhang Y, Ding L, Fu J. Downregulation of AC092894.1 promotes oxaliplatin resistance in colorectal cancer via the USP3/AR/RASGRP3 axis. BMC Med 2023; 21:132. [PMID: 37013584 PMCID: PMC10071743 DOI: 10.1186/s12916-023-02826-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/09/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Oxaliplatin resistance is a complex process and has been one of the most disadvantageous factors and indeed a confrontation in the procedure of colorectal cancer. Recently, long non-coding RNAs (lncRNAs) have emerged as novel molecules for the treatment of chemoresistance, but the specific molecular mechanisms mediated by them are poorly understood. METHODS The lncRNAs associated with oxaliplatin resistance were screened by microarray. lncRNA effects on oxaliplatin chemoresistance were then verified by gain- and loss-of-function experiments. Finally, the potential mechanism of AC092894.1 was explored by RNA pull-down, RIP, and Co-IP experiments. RESULTS AC092894.1 representation has been demonstrated to be drastically downregulated throughout oxaliplatin-induced drug-resistant CRC cells. In vivo and in vitro experiments revealed that AC092894.1 functions to reverse chemoresistance. Studies on the mechanism suggested that AC092894.1 served as a scaffold molecule that mediated the de-ubiquitination of AR through USP3, thereby increasing the transcription of RASGRP3. Finally, sustained activation of the MAPK signaling pathway induced apoptosis in CRC cells. CONCLUSIONS In conclusion, this study identified AC092894.1 as a suppressor of CRC chemoresistance and revealed the idea that targeting the AC092894.1/USP3/AR/RASGRP3 signaling axis is a novel option for the treatment of oxaliplatin resistance.
Collapse
Affiliation(s)
- Zhijian Zheng
- Department of Medical Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
- Department of Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Ming Wu
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Hongyan Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wenxia Xu
- Department of Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Mengxiang Yang
- Department of Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Kailing Pan
- Department of Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Yuqi Ni
- Department of Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Ting Jiang
- Department of Nuclear Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Hongjuan Zheng
- Department of Medical Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Xiayun Jin
- Department of Medical Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Yanfei Zhang
- Department of Pathology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Linchao Ding
- Department of Scientific Research, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China.
| | - Jianfei Fu
- Department of Medical Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China.
| |
Collapse
|
32
|
Lee CC, Lee AW, Wei PL, Liu YS, Chang YJ, Huang CY. In silico analysis to identify miR-1271-5p/PLCB4 (phospholipase C Beta 4) axis mediated oxaliplatin resistance in metastatic colorectal cancer. Sci Rep 2023; 13:4366. [PMID: 36927770 PMCID: PMC10020571 DOI: 10.1038/s41598-023-31331-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Oxaliplatin (OXA) is the first-line chemotherapy drug for metastatic colorectal cancer (mCRC), and the emergence of drug resistance is a major clinical challenge. Although there have been numerous studies on OXA resistance, but its underlying molecular mechanisms are still unclear. This study aims to identify key regulatory genes and pathways associated with OXA resistance. The Gene Expression Omnibus (GEO) GSE42387 dataset containing gene expression profiles of parental and OXA-resistant LoVo cells was applied to explore potential targets. GEO2R, STRING, CytoNCA (a plug-in of Cytoscape), and DAVID were used to analyze differentially expressed genes (DEGs), protein-protein interactions (PPIs), hub genes in PPIs, and gene ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. R2 online platform was used to run a survival analysis of validated hub genes enriched in KEGG pathways. The ENCORI database predicted microRNAs for candidate genes. A survival analysis of those genes was performed, and validated using the OncoLnc database. In addition, the 'clusterProfiler' package in R was used to perform gene set enrichment analysis (GSEA). We identified 395 DEGs, among which 155 were upregulated and 240 were downregulated. In total, 95 DEGs were screened as hub genes after constructing the PPI networks. Twelve GO terms and three KEGG pathways (steroid hormone biosynthesis, malaria, and pathways in cancer) were identified as being significant in the enrichment analysis of hub genes. Twenty-one hub genes enriched in KEGG pathways were defined as key genes. Among them AKT3, phospholipase C Beta 4 (PLCB4), and TGFB1 were identified as OXA-resistance genes through the survival analysis. High expressions of AKT3 and TGFB1 were each associated with a poor prognosis, and lower expression of PLCB4 was correlated with worse survival. Further, high levels of hsa-miR-1271-5p, which potentially targets PLCB4, were associated with poor overall survival in patients with CRC. Finally, we found that PLCB4 low expression was associated with MAPK signaling pathway and VEGF signaling pathway in CRC. Our results demonstrated that hsa-miR-1271-5p/PLCB4 in the pathway in cancer could be a new potential therapeutic target for mCRC with OXA resistance.
Collapse
Affiliation(s)
- Cheng-Chin Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Ai-Wei Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC. .,Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC.
| | - Po-Li Wei
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC.,Division of Colorectal Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan, ROC.,Cancer Research Center and Translational Laboratory, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan, ROC.,Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan, ROC
| | - Yi-Shin Liu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Yu-Jia Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC. .,Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan, ROC. .,Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan, ROC.
| | - Chien-Yu Huang
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC. .,Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC.
| |
Collapse
|
33
|
Yang IP, Yip KL, Chang YT, Chen YC, Huang CW, Tsai HL, Yeh YS, Wang JY. MicroRNAs as Predictive Biomarkers in Patients with Colorectal Cancer Receiving Chemotherapy or Chemoradiotherapy: A Narrative Literature Review. Cancers (Basel) 2023; 15:cancers15051358. [PMID: 36900159 PMCID: PMC10000071 DOI: 10.3390/cancers15051358] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/12/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies and is associated with high mortality rates worldwide. The underlying mechanism of tumorigenesis in CRC is complex, involving genetic, lifestyle-related, and environmental factors. Although radical resection with adjuvant FOLFOX (5-fluorouracil, leucovorin, and oxaliplatin) chemotherapy and neoadjuvant chemoradiotherapy have remained mainstays of treatment for patients with stage III CRC and locally advanced rectal cancer, respectively, the oncological outcomes of these treatments are often unsatisfactory. To improve patients' chances of survival, researchers are actively searching for new biomarkers to facilitate the development of more effective treatment strategies for CRC and metastatic CRC (mCRC). MicroRNAs (miRs), small, single-stranded, noncoding RNAs, can post-transcriptionally regulate mRNA translation and trigger mRNA degradation. Recent studies have documented aberrant miR levels in patients with CRC or mCRC, and some miRs are reportedly associated with chemoresistance or radioresistance in CRC. Herein, we present a narrative review of the literature on the roles of oncogenic miRs (oncomiRs) and tumor suppressor miRs (anti-oncomiRs), some of which can be used to predict the responses of patients with CRC to chemotherapy or chemoradiotherapy. Moreover, miRs may serve as potential therapeutic targets because their functions can be manipulated using synthetic antagonists and miR mimics.
Collapse
Affiliation(s)
- I-Ping Yang
- Department of Nursing, Shu-Zen College of Medicine and Management, Kaohsiung 82144, Taiwan
| | - Kwan-Ling Yip
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yu-Tang Chang
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Division of Pediatric Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yen-Cheng Chen
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Wen Huang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hsiang-Lin Tsai
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yung-Sung Yeh
- Division of Trauma and Surgical Critical Care, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Emergency Medicine, Faculty of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Graduate Institute of Injury Prevention and Control, College of Public Health, Taipei Medical University, Taipei 11031, Taiwan
| | - Jaw-Yuan Wang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Pingtung Hospital, Ministry of Health and Welfare, Pingtung 90054, Taiwan
- Correspondence:
| |
Collapse
|
34
|
Tang YL, Li DD, Duan JY, Sheng LM, Wang X. Resistance to targeted therapy in metastatic colorectal cancer: Current status and new developments. World J Gastroenterol 2023; 29:926-948. [PMID: 36844139 PMCID: PMC9950860 DOI: 10.3748/wjg.v29.i6.926] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/24/2022] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most lethal and common malignancies in the world. Chemotherapy has been the conventional treatment for metastatic CRC (mCRC) patients. However, the effects of chemotherapy have been unsatisfactory. With the advent of targeted therapy, the survival of patients with CRC have been prolonged. Over the past 20 years, targeted therapy for CRC has achieved substantial progress. However, targeted therapy has the same challenge of drug resistance as chemotherapy. Consequently, exploring the resistance mechanism and finding strategies to address the resistance to targeted therapy, along with searching for novel effective regimens, is a constant challenge in the mCRC treatment, and it is also a hot research topic. In this review, we focus on the current status on resistance to existing targeted therapies in mCRC and discuss future developments.
Collapse
Affiliation(s)
- Yuan-Ling Tang
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Dan-Dan Li
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jia-Yu Duan
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Lei-Ming Sheng
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xin Wang
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
35
|
Dinu IM, Mihăilă M, Diculescu MM, Croitoru VM, Turcu-Stiolica A, Bogdan D, Miron MI, Lungulescu CV, Alexandrescu ST, Dumitrașcu T, Buică F, Luca IN, Lungulescu C, Negulescu MC, Gramaticu IM, Cazacu IM, Croitoru AE. Bevacizumab Treatment for Metastatic Colorectal Cancer in Real-World Clinical Practice. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:350. [PMID: 36837551 PMCID: PMC9963555 DOI: 10.3390/medicina59020350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/02/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023]
Abstract
Background and Objectives: Colorectal cancer (CRC) is a leading cause of cancer-related mortality and morbidity worldwide. Bevacizumab was approved for the treatment of metastatic colorectal cancer (mCRC) based on favorable benefit-risk assessments from randomized controlled trials, but evidence on its use in the real-world setting is limited. The aim of the current study is to evaluate the outcomes and safety profile of bevacizumab in mCRC in a real-world setting in Romania. Patients and Methods: This was an observational, retrospective, multicentric, cohort study conducted in Romania that included patients with mCRC treated with bevacizumab as part of routine clinical practice. Study endpoints were progression-free survival, overall survival, adverse events, and patterns of bevacizumab use. Results: A total of 554 patients were included in the study between January 2008 and December 2018. A total of 392 patients (71%) received bevacizumab in the first line and 162 patients (29%) in the second line. Bevacizumab was mostly combined with a capecitabine/oxaliplatin chemotherapy regimen (31.6%). The median PFS for patients treated with bevacizumab was 8.4 months (interquartile range [IQR], 4.7-15.1 months) in the first line and 6.6 months (IQR, 3.8-12.3 months) in the second line. The median OS was 17.7 months (IQR, 9.3-30.6 months) in the first line and 13.5 months (IQR, 6.7-25.2 months) in the second line. Primary tumor resection was associated with a longer PFS and OS. The safety profile of bevacizumab combined with chemotherapy was similar to other observational studies in mCRC. Conclusions: The safety profile of bevacizumab was generally as expected. Although the PFS was generally similar to that reported in other studies, the OS was shorter, probably due to the less frequent use of bevacizumab after disease progression and the baseline patient characteristics. Patients with mCRC treated with bevacizumab who underwent resection of the primary tumor had a higher OS compared to patients with an unresected primary tumor.
Collapse
Affiliation(s)
- Ioana Mihaela Dinu
- Department of Oncology, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Mariana Mihăilă
- Department of Internal Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Mircea Mihai Diculescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Gastroenterology, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Vlad Mihai Croitoru
- Department of Oncology, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Adina Turcu-Stiolica
- Department of Pharmacoeconomics, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Diana Bogdan
- Department of Oncology, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Monica Ionela Miron
- Department of Oncology, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Cristian Virgil Lungulescu
- Department of Oncology, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania
- Department of Oncology, County Clinical Emergency Hospital, 200642 Craiova, Romania
| | - Sorin Tiberiu Alexandrescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Surgery, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Traian Dumitrașcu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Surgery, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Florina Buică
- Department of Oncology, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania
| | - Ioana Niculina Luca
- Department of Oncology, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Cristina Lungulescu
- Department of Oncology, County Clinical Emergency Hospital, 200642 Craiova, Romania
| | | | | | - Irina Mihaela Cazacu
- Department of Oncology, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Adina Emilia Croitoru
- Department of Oncology, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Department of Surgery, Fundeni Clinical Institute, 022328 Bucharest, Romania
| |
Collapse
|
36
|
Park KT, Jo H, Kim B, Kim W. Red Ginger Extract Prevents the Development of Oxaliplatin-Induced Neuropathic Pain by Inhibiting the Spinal Noradrenergic System in Mice. Biomedicines 2023; 11:432. [PMID: 36830967 PMCID: PMC9953630 DOI: 10.3390/biomedicines11020432] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023] Open
Abstract
Oxaliplatin is a well-known chemotherapeutic drug that is widely used to treat colorectal cancer. However, it can induce acute side effects in up to 90% of patients. Serotonin and norepinephrine reuptake inhibitors (SNRIs) are used as first-choice drugs; however, even SNRIs are known to be effective only in treatment and not for prevention. Therefore, finding a drug that can prevent the development of cold and mechanical forms of allodynia induced by oxaliplatin is needed. This study demonstrated that multiple oral administrations of 100 mg/kg and 300 mg/kg of red ginger extract could significantly prevent pain development in mice. The role of the noradrenergic system was investigated as an underlying mechanism of action. Both the spinal α1- and α2-adrenergic receptors were significantly downregulated after treatment. Furthermore, the noradrenaline levels in the serum and spinal cord were upregulated and downregulated after treatment with paclitaxel and red ginger, respectively. As the active sub-component of red ginger, ginsenoside Rg3 (Rg3) was identified and quantified using HPLC. Moreover, multiple intraperitoneal injections of Rg3 prevented the development of pain in paclitaxel-treated mice, suggesting that RG3 may induce the effect of red ginger extract.
Collapse
Affiliation(s)
- Keun-Tae Park
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Heejoon Jo
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Bonglee Kim
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Woojin Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
37
|
de Back T, Nijskens I, Schafrat P, Chalabi M, Kazemier G, Vermeulen L, Sommeijer D. Evaluation of Systemic Treatments of Small Intestinal Adenocarcinomas: A Systematic Review and Meta-analysis. JAMA Netw Open 2023; 6:e230631. [PMID: 36826817 PMCID: PMC9958532 DOI: 10.1001/jamanetworkopen.2023.0631] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 01/09/2023] [Indexed: 02/25/2023] Open
Abstract
Importance Although small intestinal adenocarcinomas (SIAs) are rare, they have a poor prognosis, and the optimal treatment strategies are largely unknown. Because of the lack of high-quality evidence, guidelines for colorectal cancer are often followed in the treatment of SIAs. Objective To review the current evidence regarding survival benefit of systemic therapies, including chemotherapy, targeted agents, and immunotherapy, for patients with SIAs. Data Sources Following the Preferred Reporting Items for Systematic Reviews and Meta-analyses, MEDLINE and Embase were searched for articles published from January 1, 2005, until June 1, 2022. Study Selection Retrospective cohort studies and prospective phase 2 or 3 trials describing survival after systemic therapies for patients with SIAs were eligible for inclusion. Assessment of study eligibility was blinded and performed by 3 reviewers. Data Extraction and Synthesis The reviewers independently extracted data. Random effects, inverse variance, pairwise meta-analyses were performed. Main Outcomes and Measures Primary outcomes were overall survival (OS) and progression-free survival (PFS) of patients with SIAs after systemic therapies. Measures of interest included hazard ratios for survival and median survival times. Results Overall, 57 retrospective cohort and phase 2 studies of 35 176 patients were included. Adjuvant chemotherapy, generally fluoropyrimidine-based, was associated with increased OS in stage I to III SIAs (hazard ratio [HR], 0.60; 95% CI, 0.53-0.68), especially in stage III tumors (HR, 0.55; 95% CI, 0.48-0.64), irrespective of tumor localization. Palliative chemotherapy was also associated with an OS benefit (HR, 0.48; 95% CI, 0.40-0.58). Fluoropyrimidine-oxaliplatin combinations were superior to other regimens (OS: HR, 0.54; 95% CI, 0.30-0.99; PFS: HR, 0.46; 95% CI, 0.30-0.71). Furthermore, bevacizumab added to chemotherapy compared with chemotherapy alone was associated with significantly prolonged PFS (HR, 0.62; 95% CI, 0.43-0.89). Immunotherapy showed a 50% overall response rate in previously treated defective mismatch repair tumors. Conclusions and Relevance In this systematic review and meta-analysis, adjuvant and palliative chemotherapy were both associated with improved survival of patients with SIAs, especially fluoropyrimidine-based regimens and fluoropyrimidine-oxaliplatin combinations. Adding bevacizumab to chemotherapy appears to prolong PFS and deserves further investigation. Immunotherapy seems beneficial and should be considered for patients with defective mismatch repair tumors. International collaborations should be undertaken to confirm and improve efficacy of systemic therapies for patients with SIAs.
Collapse
Affiliation(s)
- Tim de Back
- Center for Experimental and Molecular Medicine, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, the Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Isabelle Nijskens
- Department of Internal Medicine, Flevohospital, Almere, the Netherlands
| | - Pascale Schafrat
- Center for Experimental and Molecular Medicine, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, the Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Myriam Chalabi
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Geert Kazemier
- Department of Pathology, Amsterdam UMC, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Surgery, Amsterdam UMC, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Louis Vermeulen
- Center for Experimental and Molecular Medicine, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, the Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands
- Department of Medical Oncology, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
| | - Dirkje Sommeijer
- Center for Experimental and Molecular Medicine, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands
- Department of Internal Medicine, Flevohospital, Almere, the Netherlands
- Department of Medical Oncology, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
38
|
Zhou J, Jiang X, Xia HA, Hobbs BP, Wei P. Landmark mediation survival analysis using longitudinal surrogate. Front Oncol 2023; 12:999324. [PMID: 36733365 PMCID: PMC9887328 DOI: 10.3389/fonc.2022.999324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 12/14/2022] [Indexed: 01/18/2023] Open
Abstract
Clinical cancer trials are designed to collect radiographic measurements of each patient's baseline and residual tumor burden at regular intervals over the course of study. For solid tumors, the extent of reduction in tumor size following treatment is used as a measure of a drug's antitumor activity. Statistical estimation of treatment efficacy routinely reduce the longitudinal assessment of tumor burden to a binary outcome describing the presence versus absence of an objective tumor response as defined by RECIST criteria. The objective response rate (ORR) is the predominate method for evaluating an experimental therapy in a single-arm trial. Additionally, ORR is routinely compared against a control therapy in phase III randomized controlled trials. The longitudinal assessments of tumor burden are seldom integrated into a formal statistical model, nor integrated into mediation analysis to characterize the relationships among treatment, residual tumor burden, and survival. This article presents a frameworkfor landmark mediation survival analyses devised to incorporate longitudinal assessment of tumor burden. R 2 effect-size measures are developed to quantify the survival treatment mediation effects using longitudinal predictors. Analyses are demonstrated with applications to two colorectal cancer trials. Survival prediction is compared in the presence versus absence of longitudinal analysis. Simulation studies elucidate settings wherein patterns of tumor burden dynamics require longitudinal analysis.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Biostatistics and Pharmacometrics, Neuroscience Global Drug Development, Novartis, East Hanover, NJ, United States
| | - Xun Jiang
- Center for Design and Analysis, Amgen, Thousand Oaks, CA, United States
| | - H. Amy Xia
- Center for Design and Analysis, Amgen, Thousand Oaks, CA, United States
| | - Brian P. Hobbs
- Department of Population Health, The University of Texas, Austin, TX, United States
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States,*Correspondence: Peng Wei,
| |
Collapse
|
39
|
Wong AHN, Ma B, Lui RN. New developments in targeted therapy for metastatic colorectal cancer. Ther Adv Med Oncol 2023; 15:17588359221148540. [PMID: 36687386 PMCID: PMC9846305 DOI: 10.1177/17588359221148540] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/14/2022] [Indexed: 01/18/2023] Open
Abstract
Colorectal cancer (CRC) is the second most lethal cancer worldwide and the prognosis of metastatic CRC (mCRC) remains poor. Recent advancements in translational research have led to the identification of several new therapeutic targets and improved the treatment outcome of patients with tumours harbouring BRAF V600E mutation, (HER2) ErBB2 alterations, NTRK gene fusions and KRAS(G12C) mutation. Improved understanding towards the mechanism of resistance to targeted therapy such as anti-epidermal growth factor receptor antibodies and the evolving role of therapeutic monitoring with circulating tumour DNA (ctDNA) has enabled the longitudinal tracking of clonal evolution during treatment and the individualization of subsequent treatments. To broaden the community-based implementation of precision oncology in directing targeted therapies for patients with gastrointestinal cancers including mCRC, the feasibility of 'Master Protocols' that utilizes ctDNA-based genotyping platforms is currently being evaluated. Such protocols encompass both observational and interventional clinical trials of novel targeted therapies conducted within a large clinical trial network. In this review, we will discuss the latest developments in targeted therapies, and therapeutic strategies for overcoming acquired drug resistance in patients with mCRC.
Collapse
Affiliation(s)
- Ambrose H. N. Wong
- Faculty of Medicine, The Chinese University of
Hong Kong, Hong Kong SAR, China
| | - Brigette Ma
- State Key Laboratory of Translational Oncology,
Sir YK Pao Centre for Cancer, Department of Clinical Oncology, Hong Kong
Cancer Institute, Hong Kong SAR, China
| | - Rashid N. Lui
- Department of Clinical Oncology, and Division
of Gastroenterology and Hepatology, Department of Medicine and Therapeutics,
Institute of Digestive Disease, The Chinese University of Hong Kong, 9/F,
Prince of Wales Hospital, Shatin, New Territories, Hong Kong SAR,
China
| |
Collapse
|
40
|
Current Targeted Therapy for Metastatic Colorectal Cancer. Int J Mol Sci 2023; 24:ijms24021702. [PMID: 36675216 PMCID: PMC9864602 DOI: 10.3390/ijms24021702] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/07/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common type of cancer and the second leading cause of cancer deaths worldwide. Surgery or surgery plus radiotherapy and/or chemotherapy for patients with metastatic CRC (mCRC) were accepted as the main therapeutic strategies until the early 2000s, when targeted drugs, like cetuximab and bevacizumab, were developed. The use of targeted drugs in clinical practice has significantly increased patients' overall survival. To date, the emergence of several types of targeted drugs has opened new possibilities and revealed new prospects for mCRC treatment. Therapeutic strategies are continually being updated to select the most suitable targeted drugs based on the results of clinical trials that are currently underway. This review discusses the up-to date molecular evidence of targeted therapy for mCRC and summarizes the Food and Drug Administration-approved targeted drugs including the results of clinical trials. We also explain their mechanisms of action and how these affect the choice of a suitable targeted therapy.
Collapse
|
41
|
D’Arca D, Severi L, Ferrari S, Dozza L, Marverti G, Magni F, Chinello C, Pagani L, Tagliazucchi L, Villani M, d’Addese G, Piga I, Conteduca V, Rossi L, Gurioli G, De Giorgi U, Losi L, Costi MP. Serum Mass Spectrometry Proteomics and Protein Set Identification in Response to FOLFOX-4 in Drug-Resistant Ovarian Carcinoma. Cancers (Basel) 2023; 15:cancers15020412. [PMID: 36672361 PMCID: PMC9856519 DOI: 10.3390/cancers15020412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Ovarian cancer is a highly lethal gynecological malignancy. Drug resistance rapidly occurs, and different therapeutic approaches are needed. So far, no biomarkers have been discovered to predict early response to therapies in the case of multi-treated ovarian cancer patients. The aim of our investigation was to identify a protein panel and the molecular pathways involved in chemotherapy response through a combination of studying proteomics and network enrichment analysis by considering a subset of samples from a clinical setting. Differential mass spectrometry studies were performed on 14 serum samples from patients with heavily pretreated platinum-resistant ovarian cancer who received the FOLFOX-4 regimen as a salvage therapy. The serum was analyzed at baseline time (T0) before FOLFOX-4 treatment, and before the second cycle of treatment (T1), with the aim of understanding if it was possible, after a first treatment cycle, to detect significant proteome changes that could be associated with patients responses to therapy. A total of 291 shared expressed proteins was identified and 12 proteins were finally selected between patients who attained partial response or no-response to chemotherapy when both response to therapy and time dependence (T0, T1) were considered in the statistical analysis. The protein panel included APOL1, GSN, GFI1, LCATL, MNA, LYVE1, ROR1, SHBG, SOD3, TEC, VPS18, and ZNF573. Using a bioinformatics network enrichment approach and metanalysis study, relationships between serum and cellular proteins were identified. An analysis of protein networks was conducted and identified at least three biological processes with functional and therapeutic significance in ovarian cancer, including lipoproteins metabolic process, structural component modulation in relation to cellular apoptosis and autophagy, and cellular oxidative stress response. Five proteins were almost independent from the network (LYVE1, ROR1, TEC, GFI1, and ZNF573). All proteins were associated with response to drug-resistant ovarian cancer resistant and were mechanistically connected to the pathways associated with cancer arrest. These results can be the basis for extending a biomarker discovery process to a clinical trial, as an early predictive tool of chemo-response to FOLFOX-4 of heavily treated ovarian cancer patients and for supporting the oncologist to continue or to interrupt the therapy.
Collapse
Affiliation(s)
- Domenico D’Arca
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy
| | - Leda Severi
- Department Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Stefania Ferrari
- Department Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Luca Dozza
- Seràgnoli Institute of Hematology, Department of Experimental, Diagnostic and Specialty Medicine, Bologna University School of Medicine, S. Orsola Malpighi Hospital, 40138 Bologna, Italy
| | - Gaetano Marverti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy
| | - Fulvio Magni
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, 20126 Vedano al Lambro, Italy
| | - Clizia Chinello
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, 20126 Vedano al Lambro, Italy
| | - Lisa Pagani
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, 20126 Vedano al Lambro, Italy
| | - Lorenzo Tagliazucchi
- Department Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
- Clinical and Experimental Medicine (CEM) Doctorate School, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy
| | - Marco Villani
- Department of Physics, Informatics and Mathematics, Modena and Reggio Emilia University, Via Campi 213/A, 41125 Modena, Italy
| | - Gianluca d’Addese
- Department of Physics, Informatics and Mathematics, Modena and Reggio Emilia University, Via Campi 213/A, 41125 Modena, Italy
| | - Isabella Piga
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, 20126 Vedano al Lambro, Italy
| | - Vincenza Conteduca
- IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), 47014 Meldola, Italy
| | - Lorena Rossi
- IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), 47014 Meldola, Italy
| | - Giorgia Gurioli
- IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), 47014 Meldola, Italy
| | - Ugo De Giorgi
- IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), 47014 Meldola, Italy
| | - Lorena Losi
- Department Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
- Correspondence: (L.L.); (M.P.C.)
| | - Maria Paola Costi
- Department Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
- Correspondence: (L.L.); (M.P.C.)
| |
Collapse
|
42
|
Foster JM, Zhang C, Rehman S, Sharma P, Alexander HR. The contemporary management of peritoneal metastasis: A journey from the cold past of treatment futility to a warm present and a bright future. CA Cancer J Clin 2023; 73:49-71. [PMID: 35969103 DOI: 10.3322/caac.21749] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/12/2022] [Accepted: 06/15/2022] [Indexed: 01/17/2023] Open
Abstract
Peritoneal metastasis (PM) is often regarded as a less frequent pattern of spread; however, collectively across all spectra of primary tumors, the consequences of PM impact a large population of patients annually. Unlike other modes of metastasis, symptoms at presentation or during the treatment course are common, representing an additional challenge in the management of PM. Early efforts with chemotherapy and incomplete surgical interventions transiently improved symptoms, but durable symptom control and survival extension were rare, which established a perspective of treatment futility for PM through most of the 20th century. Notably, the continued development of better systemic therapy combinations, optimization of cytoreductive surgery (CRS), and rigorous investigation of combining regional therapy-specifically hyperthermic intraperitoneal chemotherapy-with CRS, have resulted in more effective multimodal treatment options for patients with PM. In this article, the authors provide a comprehensive review of the data establishing the contemporary approach for tumors with a high frequency of PM, including appendix, colorectal, mesothelioma, and gastric cancers. The authors also explore the emerging role of adding hyperthermic intraperitoneal chemotherapy to the well established paradigm of CRS and systemic therapy for advanced ovarian cancer, as well as the recent clinical trials identifying the efficacy of poly(adenosine diphosphate ribose) polymerase maintenance therapy. Finally, recent data are included that explore the role of precision medicine technology in PM management that, in the future, may help further improve patient selection, identify the best systemic therapy regimens, detect actionable mutations, and identify new targets for drug development.
Collapse
Affiliation(s)
- Jason M Foster
- Division of Surgical Oncology, Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Chunmeng Zhang
- Division of Surgical Oncology, Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Shahyan Rehman
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey
| | - Prateek Sharma
- Division of Surgical Oncology, Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | |
Collapse
|
43
|
Siddique A, Bashir S, Abbas M. Pharmacogenetics of Anticancer Drugs: Clinical Response and Toxicity. Cancer Treat Res 2023; 185:141-175. [PMID: 37306909 DOI: 10.1007/978-3-031-27156-4_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Cancer is the most challenging disease for medical professionals to treat. The factors underlying the complicated situation include anticancer drug-associated toxicity, non-specific response, low therapeutic window, variable treatment outcomes, development of drug resistance, treatment complications, and cancer recurrence. The remarkable advancement in biomedical sciences and genetics, over the past few decades, however, is changing the dire situation. The discovery of gene polymorphism, gene expression, biomarkers, particular molecular targets and pathways, and drug-metabolizing enzymes have paved the way for the development and provision of targeted and individualized anticancer treatment. Pharmacogenetics is the study of genetic factors having the potential to affect clinical responses and pharmacokinetic and pharmacodynamic behaviors of drugs. This chapter emphasizes pharmacogenetics of anticancer drugs and its applications in improving treatment outcomes, selectivity, toxicity of the drugs, and discovering and developing personalized anticancer drugs and genetic methods for prediction of drug response and toxicity.
Collapse
Affiliation(s)
- Ammara Siddique
- Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Samra Bashir
- Faculty of Pharmacy, Capital University of Science and Technology, Islamabad, Pakistan.
| | - Mateen Abbas
- Faculty of Pharmacy, Capital University of Science and Technology, Islamabad, Pakistan
| |
Collapse
|
44
|
McGarrah P, Hubbard J, Novotny PJ, Branda ME, Sargent DS, Morton RF, Fuchs CS, Benson AB, Williamson SK, Findlay BP, Alberts SR, Goldberg RM, Sloan JA. Baseline Quality of Life is a Strong and Independent Prognostic Factor for Overall Survival in Metastatic Colorectal Cancer. Cancer Control 2023; 30:10732748231185047. [PMID: 37339926 PMCID: PMC10286175 DOI: 10.1177/10732748231185047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND Previous studies have established that higher baseline quality of life (QOL) scores are associated with improved survival in patients with metastatic colorectal cancer (mCRC). We examined the relationship between overall survival (OS) and baseline QOL. PATIENTS AND METHODS A total of 1 247 patients with mCRC participating in N9741 (comparing bolus 5-FU/LV, irinotecan [IFL] vs infusional 5-FU/leucovorin [LV]/oxaliplatin [FOLFOX] vs. irinotecan/oxaliplatin [IROX]) provided data at baseline on overall QOL using a single-item linear analogue self-assessment (LASA) 0-100 point scale. The association of OS according to clinically deficient (defined as CD-QOL, score 0-50) vs not clinically deficient (nCD-QOL, score 51-100) baseline QOL scores was tested. A multivariable analysis using Cox proportional hazards modeling was performed to adjust for the effects of multiple baseline factors. An exploratory analysis was performed evaluating OS according to baseline QOL status among patients who did or did not receive second-line therapy. RESULTS Baseline QOL was a strong predictor of OS for the whole cohort (CD-QOL vs nCD-QOL: 11.2 months vs 18.4 months, P < .0001), and in each arm IFL 12.4 vs 15.1 months, FOLFOX 11.1 months vs 20.6 months, and IROX 8.9 months vs 18.1 months. Baseline QOL was associated with baseline performance status (PS) (P < .0001). After adjusting for PS and treatment arm, baseline QOL was still associated with OS (P = .017). CONCLUSIONS Baseline QOL is an independent prognostic factor for OS in patients with mCRC. The demonstration that patient-assessed QOL and PS are independent prognostic indicators suggests that these assessments provide important complementary prognostic information.
Collapse
Affiliation(s)
| | - Joleen Hubbard
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Paul J. Novotny
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, USA
| | - Megan E. Branda
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, USA
| | - Daniel S. Sargent
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, USA
| | - Roscoe F. Morton
- North Central Cancer Treatment Group, Iowa Oncology Research Association, Des Moines, IA, USA
| | | | - Al B. Benson
- Division of Hematology and Medical Oncology, Northwestern University, Chicago, IL, USA; and ECOG-ACRIN Cancer Research Group, Philadelphia, PA, USA
| | - Stephen K. Williamson
- SWOG Cancer Research Network, Division of Hematology and Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | | | | | | | - Jeff A. Sloan
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
45
|
Shiraishi T, Ogawa H, Shioi I, Ozawa N, Osone K, Okada T, Sohda M, Shirabe K, Saeki H. Differences in prognosis and underuse of adjuvant chemotherapy between elderly and non-elderly patients in stage III colorectal cancer. Ann Gastroenterol Surg 2023; 7:91-101. [PMID: 36643370 PMCID: PMC9831896 DOI: 10.1002/ags3.12604] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/06/2022] [Indexed: 01/18/2023] Open
Abstract
Aim We aimed to clarify the use of adjuvant chemotherapy and the prognosis of elderly colorectal cancer patients compared with non-elderly patients, and the usefulness of sarcopenia as an indicator for the introduction and completion of adjuvant chemotherapy. Methods Between 2013 and 2021, 215 patients with pStage III disease were included. We investigated perioperative clinicopathological factors, adjuvant chemotherapy details, and prognosis. Preoperative sarcopenia status was evaluated using computed tomography images. Elderly patients were defined as those aged ≥70 years. Results We included 121 (56.3%) and 94 (43.7%) non-elderly and elderly patients, respectively. Among the elderly patients, 47 had sarcopenia. There were no significant differences in the incompletion rate of adjuvant chemotherapy between elderly and non-elderly patients (27.1%/16.2%, P = 0.119). The most common reason for the discontinuation of adjuvant chemotherapy was side effects, regardless of age. The respective 3-year-disease free survival of patients with no/completed/incomplete adjuvant chemotherapy were 65.5%, 80.2%, and 57.7% for non-elderly patients (P = 0.045) and 73.4%, 70.6%, and 71.6% for elderly patients (P = 0.924). The number of elderly patients with sarcopenia was significantly higher in patients without adjuvant chemotherapy (P = 0.004) and those with incomplete adjuvant chemotherapy (P = 0.004). The 3-year-disease free survival of elderly sarcopenic patients without and with adjuvant chemotherapy were 78.3% and 59.2%, respectively (P = 0.833). Conclusion Elderly patients did not show a benefit of adjuvant chemotherapy regardless of whether they had completed adjuvant chemotherapy, unlike non-elderly patients. Moreover, the evaluation of preoperative sarcopenia in elderly colorectal cancer patients may be useful in determining the indication for adjuvant chemotherapy.
Collapse
Affiliation(s)
- Takuya Shiraishi
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Hiroomi Ogawa
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Ikuma Shioi
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Naoya Ozawa
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Katsuya Osone
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Takuhisa Okada
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Makoto Sohda
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Ken Shirabe
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Hiroshi Saeki
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| |
Collapse
|
46
|
Islam MR, Akash S, Rahman MM, Nowrin FT, Akter T, Shohag S, Rauf A, Aljohani AS, Simal-Gandara J. Colon cancer and colorectal cancer: Prevention and treatment by potential natural products. Chem Biol Interact 2022; 368:110170. [DOI: 10.1016/j.cbi.2022.110170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/24/2022] [Accepted: 09/03/2022] [Indexed: 11/29/2022]
|
47
|
Argilés G, Mulet N, Valladares-Ayerbes M, Viéitez JM, Grávalos C, García-Alfonso P, Santos C, Tobeña M, García-Paredes B, Benavides M, Cano MT, Loupakis F, Rodríguez-Garrote M, Rivera F, Goldberg RM, Cremolini C, Bennouna J, Ciardiello F, Tabernero JM, Aranda E, Argilés G, Tabernero J. A randomised phase 2 study comparing different dose approaches of induction treatment of regorafenib in previously treated metastatic colorectal cancer patients (REARRANGE trial). Eur J Cancer 2022; 177:154-163. [PMID: 36335783 DOI: 10.1016/j.ejca.2022.09.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 02/08/2023]
Abstract
PURPOSE The purpose of this article is to evaluate the safety of two regorafenib dose-escalation approaches in refractory metastatic colorectal cancer (mCRC) patients. PATIENTS AND METHODS Patients with mCRC and progression during or within 3 months following their last standard chemotherapy regimen were randomised to receive the approved dose of regorafenib of 160 mg QD (arm A) or 120 mg QD (arm B) administered as 3 weeks of treatment followed by 1 week off, or 160 mg QD 1 week on/1 week off (arm C). The primary end-point was the percentage of patients with G3/G4 treatment-related adverse events (AEs) in each arm. RESULTS There were 299 patients randomly assigned to arm A (n = 101), arm B (n = 99), or arm C (n = 99); 297 initiated treatments (arm A n = 100, arm B n = 98, arm C n = 99: population for safety analyses). G3/4 treatment-related AEs occurred in 60%, 55%, and 54% of patients in arms A, B, and C, respectively. The most common G3/4 AEs were hypertension (19, 12, and 20 patients), fatigue (20, 14, and 15 patients), hypokalemia (11, 7, and 10 patients), and hand-foot skin reaction (8, 7, and 3 patients). Median overall survival was 7.4 (IQR 4.0-13.7) months in arm A, 8.6 (IQR 3.8-13.4) in arm B, and 7.1 (IQR 4.4-12.4) in arm C. CONCLUSIONS The alternative regorafenib dosing schedules were feasible and safe in patients with mCRC who had been previously treated with standard therapy. There was a higher numerical improvement on the most clinically relevant AEs in the intermittent dosing arm, particularly during the relevant first two cycles. GOV IDENTIFIER NCT02835924.
Collapse
Affiliation(s)
- Guillem Argilés
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), CIBERONC, Barcelona, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain; Memorial Sloan Kettering Cancer Center, New York, USA.
| | - Nuria Mulet
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), CIBERONC, Barcelona, Spain
| | | | - José M Viéitez
- Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | | | - Cristina Santos
- Institute Català d'Oncologia (ICO), Duran i Reynals Hospital - ONCOBELL, CIBERONC, Barcelona, Spain
| | - María Tobeña
- Santa Creu i Sant Pau Hospital, Barcelona, Spain
| | - Beatriz García-Paredes
- Clínico San Carlos Hospital, Instituto de Investigación Hospital Clinico San Carlos (IdISSC) CIBERONC, Madrid, Spain
| | | | - María T Cano
- IMIBIC, Reina Sofía Hospital, University of Córdoba, CIBERONC, Instituto de Salud Carlos III, Spain
| | | | | | - Fernando Rivera
- University Hospital Marqués de Valdecilla (IDIVAL) Santander, Spain
| | | | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Jaafar Bennouna
- University Hospital of Nantes, Digestive Oncology, Nantes, France
| | | | - Josep M Tabernero
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), CIBERONC, Barcelona, Spain
| | | | - Guillem Argilés
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), CIBERONC, Barcelona, Spain
| | - Josep Tabernero
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), CIBERONC, Barcelona, Spain
| |
Collapse
|
48
|
Wu P, Pan X, Lu K, Gu N. Screening prognostic genes related to leucovorin, fluorouracil, and irinotecan treatment sensitivity by performing co-expression network analysis for colon cancer. Front Genet 2022; 13:928356. [DOI: 10.3389/fgene.2022.928356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 11/11/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Colon cancer is one of the most common malignant tumors in the world. FOLFIRI (leucovorin, fluorouracil, and irinotecan) is a common combination in chemotherapy regimens. However, insensitivity to FOLFIRI is an important factor in the effectiveness of the treatment for advanced colon cancer. Our study aimed to explore precise molecular targets associated with chemotherapy responses in colon cancer.Methods: Gene expression profiles of 21 patients with advanced colorectal cancer who received chemotherapy based on FOLFIRI were obtained from the Gene Expression Omnibus (GEO) database. The gene co-expression network was constructed by the weighted gene co-expression network analysis (WGCNA) and functional gene modules were screened out. Clinical phenotypic correlation analysis was used to identify key gene modules. Gene Ontology and pathway enrichment analysis were used to screen enriched genes in key modules. Protein–protein interaction (PPI) analysis was used to screen out key node genes. Based on the Gene Expression Profiling Interactive Analysis (GEPIA) database, the correlation between the expression levels of these genes and the overall survival (OS) and disease-free survival (DFS) of colon cancer patients was investigated, and the hub genes were screened out. Immunohistochemistry of candidate hub genes was identified using the Human Protein Atlas database. Finally, clinical information and RNA sequencing data of colon cancer were obtained from The Cancer Genome Atlas project database (TCGA), the GEPIA, and the Human Atlas databases for validation.Results: The WGCNA revealed that three hub genes were closely related to chemotherapy insensitivity of colon cancer: AEBP1, BGN, and TAGLN. The protein expression levels of AEBP1, BGN, and TAGLN in tumor tissues were higher than those in normal tissues. In addition, the gene expression levels of AEBP1, BGN, and TAGLN were negatively correlated with OS and DFS in colon cancer patients. Therefore, AEBP1, BGN, and TAGLN have been identified as potential biomarkers related to the response to FOLFIRI treatment of colon cancer.Conclusion: We found that AEBP1, BGN, and TAGLN, as potential predictive biomarkers, may play an important role in the response to FOLFIRI treatment of colon cancer and as a precise molecular target associated with chemotherapy response in colon cancer.
Collapse
|
49
|
Wang M, Xiang Y, Wang R, Zhang L, Zhang H, Chen H, Luan X, Chen L. Dihydrotanshinone I Inhibits the Proliferation and Growth of Oxaliplatin-Resistant Human HCT116 Colorectal Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227774. [PMID: 36431875 PMCID: PMC9692243 DOI: 10.3390/molecules27227774] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022]
Abstract
Oxaliplatin (OXA) is a first-line chemotherapeutic drug for the treatment of colorectal cancer (CRC), but acquired drug resistance becomes the main cause of treatment failure. Increasing evidence has shown that some natural components may serve as chemoresistant sensitizers. In this study, we discovered Dihydrotanshinone I (DHTS) through virtual screening using a ligand-based method, and explored its inhibitory effects and the mechanism on OXA-resistant CRC in vitro and in vivo. The results showed that DHTS could effectively inhibit the proliferation of HCT116 and HCT116/OXA resistant cells. DHTS-induced cell apoptosis blocked cell cycle in S and G2/M phases, and enhanced DNA damage of HCT116/OXA cells in a concentration-dependent manner. DHTS also exhibited the obvious inhibition of tumor growth in the HCT116/OXA xenograft model. Mechanistically, DHTS could downregulate the expression of Src homology 2 structural domain protein tyrosine phosphatase (SHP2) and Wnt/β-catenin, as well as conventional drug resistance and apoptosis-related proteins such as multidrug resistance associated proteins (MRP1), P-glycoprotein (P-gp), Bcl-2, and Bcl-xL. Thus, DHTS markedly induces cell apoptosis and inhibits tumor growth in OXA-resistant HCT116 CRC mice models, which can be used as a novel lead compound against OXA-resistant CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xin Luan
- Correspondence: (X.L.); (L.C.); Tel./Fax: +86-21-51322428 (X.L.); +86-21-51322720 (L.C.)
| | - Lili Chen
- Correspondence: (X.L.); (L.C.); Tel./Fax: +86-21-51322428 (X.L.); +86-21-51322720 (L.C.)
| |
Collapse
|
50
|
Yang W, Zhao Y, Ge Q, Wang X, Jing Y, Zhao J, Liu G, Huang H, Cheng F, Wang X, Ye Y, Song W, Liu X, Du J, Sheng J, Cao X. Genetic mutation and tumor microbiota determine heterogenicity of tumor immune signature: Evidence from gastric and colorectal synchronous cancers. Front Immunol 2022; 13:947080. [PMID: 36420271 PMCID: PMC9676241 DOI: 10.3389/fimmu.2022.947080] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/23/2022] [Indexed: 01/11/2024] Open
Abstract
Both colorectal and gastric cancer are lethal solid-tumor malignancies, leading to the majority of cancer-associated deaths worldwide. Although colorectal cancer (CRC) and gastric cancer (GC) share many similarities, the prognosis and drug response of CRC and GC are different. However, determinants for such differences have not been elucidated. To avoid genetic background variance, we performed multi-omics analysis, including single-cell RNA sequencing, whole-exome sequencing, and microbiome sequencing, to dissect the tumor immune signature of synchronous primary tumors of GC and CRC. We found that cellular components of juxta-tumoral sites were quite similar, while tumoral cellular components were specific to the tumoral sites. In addition, the mutational landscape and microbiome contributed to the distinct TME cellular components. Overall, we found that different prognoses and drug responses of GC and CRC were mainly due to the distinct TME determined by mutational landscape and microbiome components.
Collapse
Affiliation(s)
- Weili Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaxing Zhao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University Cancer Center, Zhejiang University, Hangzhou, China
| | - Qiongxiang Ge
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Traditional Chinese Medicine (TCM), Hangzhou, China
| | - Xiaoli Wang
- Department of Hepato-Gastroenterology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Yang Jing
- Department of Hepato-Gastroenterology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Jingwen Zhao
- Department of Hepato-Gastroenterology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Gang Liu
- Department of Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - He Huang
- Frontiers Science Center for Synthetic Biology, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Fei Cheng
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxi Wang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yulin Ye
- Department of Hepato-Gastroenterology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Wenjing Song
- Department of Pathology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Xinjuan Liu
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Juan Du
- Department of Gastroenterology, First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jianpeng Sheng
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University Cancer Center, Zhejiang University, Hangzhou, China
| | - Xiaocang Cao
- Department of Hepato-Gastroenterology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|