Published online Nov 21, 2025. doi: 10.3748/wjg.v31.i43.111433
Revised: July 28, 2025
Accepted: October 9, 2025
Published online: November 21, 2025
Processing time: 144 Days and 5.8 Hours
Pancreatic cancer is still one of the neoplasms with the worst prognosis. Late pre
Core Tip: Pancreatic cancer is still one of the neoplasms with the worst prognosis. Late presentation at an unresectable or metastatic stage precluding surgery, aggressive biology, and resistance to antiblastic drugs make this disease a fearsome challenge. This review aims to investigate new strategies for early diagnosis and new therapeutic hopes for improving the still tragic outcomes.
- Citation: Tonini V, Zanni M. Pancreatic cancer in 2025: Have we found a solution? World J Gastroenterol 2025; 31(43): 111433
- URL: https://www.wjgnet.com/1007-9327/full/v31/i43/111433.htm
- DOI: https://dx.doi.org/10.3748/wjg.v31.i43.111433
Pancreatic cancer is one of the deadliest malignancies and poses a significant global health challenge. It ranks 12th in incidence and 6th in cumulative mortality[1]. In the Western world it is the third leading cause of cancer death[2]. Despite advances in healthcare, survival rates have seen little improvement[3]. Retroperitoneal localization, nonspecific symptoms in early stages, and its aggressive biology contribute to the high percentage of patients (51%) presenting with metastatic disease at diagnosis. Additionally, the limited treatment options is due to its desmoplastic and chemoresistant nature[4]. Pancreatic ductal adenocarcinoma (PDAC) accounts for the majority (90%) of pancreatic neoplasms[4]. The 5-year overall survival (OS) rate for pancreatic cancer is 11%, while the cancer-specific survival rate is 13%[3]. The 5-year survival rate varies greatly depending on the stage of the disease at diagnosis, ranging from 44% in patients with lo
In high human development index countries such as Europe, North America, Australia/New Zealand, and Japan, the incidence of PDAC is highest, ranging from 7.9 to 9.9 per 100000 people. Conversely, Africa, Central America, and South Asia have the lowest incidence, between 1.5 and 4.6 per 100000 people[7,8]. This difference may be due to the higher pre
Several studies have examined diabetes as a risk factor. It is well established that diabetes is associated with an increased risk of PDAC and that PDAC can have a diabetogenic effect, with half of PDAC patients having diabetes[15,16]. In the first year after the diagnosis of type 2 diabetes, the risk of also being diagnosed with PDAC is 14-15 times higher than in the nondiabetic population. The risk decreases in the second year to 3.5-5.4 times and stabilizes at about 3 times[17]. Prediabetes is associated with a 40% increase in pancreatic cancer risk[18]. Type 2 diabetes significantly increases the overall annual incidence rate of PDAC compared with the general population, with a standardized incidence ratio of 1.54[19,20]. A recent systematic review suggested that new-onset diabetes, especially when associated with advanced age, a family history of pancreatic cancer, a personal history of gallstones or pancreatitis, and weight loss, is strongly correlated with PDAC and could be used as a basis for further screening strategies[21]. The association of chronic obstructive pulmonary disease or hyperuricemia with diabetes also increases the risk of pancreatic cancer[22,23]. Recent studies have also associated PDAC with conditions such as cutaneous and systemic lupus erythematosus, polycystic ovary syndrome, opium use, chronic intake of proton pump inhibitors, and chronic inflammatory bowel disease[24-29].
Heritability is suspected in 21%-36% of pancreatic cancer patients[30]. This may be due to genetic syndromes (ac
Pancreatic cancer screening in the general population is not currently recommended by guidelines[33,34]. However, it is considered in high-risk individuals (HRIs), defined as those with a lifetime risk of PDAC ≥ 5%. These include individuals with genetic syndromes or who meet criteria for FPC[31,33,34]. Besides the traditional criteria for FPC, individuals with three or more diagnoses of pancreatic cancer on the same side of the family, or with at least two affected relatives on the same side (one of whom is an FDR), are considered high risk. Screening methods include magnetic resonance imaging (MRI) or endoscopic ultrasound (EUS). MRI is better for evaluating cystic lesions, while EUS is preferred for detecting solid lesions and parenchymal changes[35]. Guidelines recommend annual screening with EUS and/or MRI[36-39]. The one-year interval is advised because nearly half of all patients develop interval PDAC at a median of 11 months in large cohorts of HRIs. Shorter intervals may be recommended if relevant lesions are detected[36]. The age to start screening depends on the individual’s estimated risk. For patients with FPC or a known genetic predisposition and one or more FDRs affected with pancreatic cancer, it is recommended to start screening at age 50 or 10 years before the age of the first affected blood relative. For those at very high risk, earlier screening is recommended, which can begin at age 40 for patients with familial atypical multiple mole melanoma or hereditary pancreatitis, and at age 35 for those with Peutz-Jeghers syndrome. There is no current consensus on when to stop screening. Discontinuation is recommended when the risk of death from causes unrelated to pancreatic cancer is higher than that of pancreatic cancer, or when patients are no longer candidates for surgical resection.
Screening programs report excellent survival outcomes, with rates ranging from 24% to 73% at 5 years. These findings contrast with sporadically diagnosed PDAC, where 5-year survival rates for all stages reach 11%-13%. Blackford et al[3] demonstrated that HRIs who undergo annual or semi-annual EUS and MRI surveillance and receive a diagnosis of pan
Carbohydrate antigen 19-9 (CA19-9) is the most validated serum tumor marker for pancreatic cancer in terms of its diagnostic, prognostic, and surveillance capabilities. However, CA19-9 has a low positive predictive value and isn’t used as a screening tool[40]. Other carbohydrate antigens have been evaluated for the early diagnosis of pancreatic cancer, but they haven’t found clinical validation. Among new diagnostic tools, circulating cell-free DNA (cfDNA), micro-RNA (miRNA), and exosomes are the most interesting. Hu et al[41], studying more than 120 cfDNA methylation biomarkers, identified a panel for PDAC detection. It includes IRX4, KCNS2, and RIMS4 and achieved a sensitivity of 86% for patients in the validation cohorts with 100% specificity. Additionally, they identified hundreds of methylated cfDNA biomarkers that differed between PDAC and hepatocarcinoma, colorectal cancer, and gastric cancer. CfDNAs are also being studied in combination with CA19-9 to improve its performance. Ben-Ami et al[42] created a panel with CA19-9, tissue inhibitor of metal protease, and cfDNA that showed higher discrimination ability than CA19-9 alone for early-stage PDACs [area under the curve (AUC): 0.86 vs 0.82].
Regarding miRNAs, the most promising panel consists of mir-5100, mir-642b-3p, and mir-125a-3p, with an AUC of 0.95, a sensitivity of 0.98, and a specificity of 0.97[43,44]. Huang et al[45] obtained similar results, reporting a signature composed of miR-132-3p, miR-30c-5p, miR-24-3p, and miR-23a-3p. It was obtained from the analysis of tissue and serum samples from 1273 participants and detected PDAC with promising accuracy (AUC = 0.971).
Excellent results were also obtained with miR-28-3p, miR-143-3p, miR-151a-3p, a panel of 2’-O-methylated miRNAs[43,46]. Exploration of exosomal protein expression has shown promising results in large cohorts[47]. Wei et al[48] and Wei et al[49] identified significantly elevated levels of the exosomal receptor erythropoietin-producing hepatocellular A2 (EphA2) in 244 pancreatic cancer patients. Indeed, the area under the receiver operating characteristic (AUROC) values for the serum exo-EphA2 diagnostic test were 0.94 and 0.92 compared with healthy controls and benign pancreatic disease, respectively. For early-stage I or II PDAC, these values decreased slightly to 0.92 and 0.90, but the combination of exo-EphA2 with CA19-9 increased the AUROC to 0.96 compared with healthy controls[48,49]. A meta-analysis on the diagnostic accuracy of exosomes in pancreatic cancer revealed that within the subset of exosomal biomarker types, the group with exosomal cell surface proteoglycan showed the highest combined sensitivity (0.96) and specificity (0.90)[50].
Extremely promising results have been obtained by analyzing biomarkers in pancreatic juice. In a recent study, Yachida et al[51] created a method to identify resectable PDAC by analyzing KRAS mutations in duodenal fluid collected during an esophagogastroduodenoscopy with stimulation of pancreatic juice secretion by secretin. They obtained an AUC of 0.934 in differentiating patients with resectable PDAC from healthy controls. In pancreatic juice, Sakaue et al[52] also detected high diagnostic potential for early-stage PDAC in Ex-miR-4516, a PDAC-specific exosomal miRNA. A multicenter prospective study evaluated 14 methylated DNA markers (MDMs) in pancreatic juice (NDRG4, BMP3, TBX15, C13orf18, PRKCB, CLEC11A, CD1D, ELMO1, IGF2BP1, RYR2, ADCY1, FER1 L4, EMX1, and LRRC4). A panel with 3 MDMs (FER1 L4, C13orf18, and BMP3) alone and in combination with CA19-9 was then studied. Methylated FER1 L4 had the highest individual AUROC of 0.83. The AUROC for the 3-MDM + plasma CA19-9 model (0.95) was higher than both the 3-MDM PJ panel (0.87) and plasma CA19-9 alone (0.91). With a specificity of 88%, the sensitivity of this model was 89% for all stages of PDAC and 83% for stage I/II PDAC[53].
The symptoms that manifest in pancreatic cancer are generally vague and nonspecific and depend on the location of the disease. Approximately 70% occur in the head of the pancreas and often present with painless jaundice and exocrine pancreatic insufficiency[5]. In contrast, tumors of the body-tail cause abdominal and back pain. In both cases, symptoms related to cachexia (loss of appetite, weight loss, fatigue) are common. The onset of diabetes or worsening of pre-existing diabetes may be a sign of PDAC. Acute pancreatitis can be a primary manifestation[5]. PDAC is accompanied by a state of hypercoagulability, which can manifest as Trousseau’s syndrome, a superficial and migratory thrombophlebitis. Skin manifestations, such as pancreatic panniculitis, may occur as paraneoplastic phenomena.
The imaging modality of choice is contrast-enhanced triphasic computed tomography (CT)[54-56]. It consists of the pancreatic, arterial, and portal venous phases. PDAC typically presents as a hypodense lesion, in stark contrast to hyperdense neuroendocrine tumors. Cancer of the head of the pancreas also often causes dilation of both the common bile duct and the main pancreatic duct, a typical feature known as the “double duct sign”[55]. However, some lesions, especially small ones, are isoattenuating and not easily identifiable[56]. It will therefore be necessary to look for indirect findings that suggest the presence of pancreatic cancer, such as dilation of the pancreatic duct with a sharp cut and pancreatic atrophy[57]. In patients with poorly defined masses or contraindications to contrast-enhanced CT, MRI or EUS may be used. MRI is also excellent for characterizing suspected liver metastases, while EUS is useful for assessing lymphovascular involvement. It is also capable of obtaining a definitive cytological or histological diagnosis. Positron emission tomography aims to rule out distant metastases in cases with a high risk of metastatic disease, for example in patients with very high serum CA19-9, regional lymphadenopathy, and large primary tumors[54-58].
In patients with suspected metastatic pancreatic cancer, a pathological report of possible metastases must be obtained[54]. In cases of localized neoplasia, a pathological report is necessary when imaging is unable to distinguish between benign and malignant disease, or in patients for whom first-line treatment is chemotherapy[54,58]. EUS with fine needle biopsy is the standard procedure[54,58]. Genetic testing for hereditary mutations is recommended for all patients diagnosed with pancreatic cancer[58]. In the case of advanced or metastatic disease, it is recommended to investigate the molecular profile to guide targeted treatment. The profile to be evaluated includes BRCA1, BRCA2, DNA mismatch repair deficiency (e.g., MLH1, MSH2, MSH6), and KRAS wild type (KRASwt) (e.g., fusion genes such as NRG and NTRK)[58].
In patients with pancreatic cancer, serum concentrations of CA19-9 and carcinoembryonic antigen (CEA), liver enzymes, and bilirubin are measured. Although the serum CA19-9 biomarker is not reliable for the definitive diagnosis of PDAC, it is valuable as an indicator of possible micro-metastatic disease, for assessing the risk of occult metastases, and for evaluating response to treatment[59].
The tumor node metastasis classification (tumor, lymph node, and metastasis) is used exclusively to assess prognosis[60]. A more practical classification, useful in everyday life to define the correct therapeutic management, divides pancreatic cancer into resectable, borderline resectable pancreatic cancer (BRPC), locally advanced pancreatic cancer (LAPC), or metastatic. Resectability is defined by anatomical (A), biological (B), and conditional (C) criteria[61,62]. Anatomical resectability is defined by the degree of contact between the tumor and adjacent vascular structures (i.e., the superior me
Surgical resection is the only potentially curative treatment, but it can only be performed in a limited number of cases, as pancreatic cancer is often diagnosed at an advanced stage. Depending on the location and extent of the tumor, a pancreatoduodenectomy, distal pancreatectomy, or total pancreatectomy may be performed. The surgical procedure involves removing the tumor with clear margins and at least twelve lymph nodes (necessary for staging). In recent years, minimally invasive procedures, particularly robotic procedures, have been implemented for pancreatic cancer, based on the assumption of a potential benefit in terms of faster recovery and the possibility for patients to proceed more quickly and more often to adjuvant therapy[63,64]. Pancreatic cancer surgery may require portomesenteric, arterial, and multivisceral vein resection to achieve radical resection. Arterial resection has historically been contraindicated due to the associated increase in morbidity and mortality. It is only considered in young patients in good general health because it can offer a more favorable prognosis than palliative treatment[64].
Open surgery remains the standard of surgical care for pancreatoduodenectomy. While the PLOT[65] and PADULAP[66] trials initially showed potential benefits for laparoscopic pancreatoduodenectomy (LPD), these results were challenged by the Dutch LEOPARD 2 study, which reported an increase in mortality in the laparoscopic group and was therefore discontinued prematurely[67]. A subsequent meta-analysis showed that the successful application of laparoscopic techniques depends on a learning curve[68]. Choi et al[69] also found an inferiority of the minimally invasive approach in terms of disease-free survival (DFS) for stage III pancreatic head tumors. Conversely, Giani et al[70] demonstrated comparability in oncologic outcomes between open and minimally invasive techniques. In fact, they found the latter was associated with better lymph node harvesting. This is especially true for the robotic technique, as confirmed by a review of 17831 pancreatoduodenectomies[71] and several single-center retrospective studies[72,73]. The EUROPA trial, a trial including 81 patients randomized to either robotic pancreatoduodenectomy (RPD) or open pancreatoduodenectomy (OPD)[74,75], found that both RPD and OPD were safe, although RPD had a higher complication rate and a longer surgical duration. When comparing laparoscopic and robotic surgery, robotic surgery often prevails. A recent study demonstrated its superiority in terms of conversion rate, blood loss, R0 resection rates, lymph node retrieval (24.2 vs 21.9), and length of hospital stay (11 days vs 13 days). Additionally, an improvement in DFS from 1 years to 3 years and OS was found in the RPD group compared with the LPD group[76].
Unlike pancreatoduodenectomy, minimally invasive distal pancreatectomy (MIDP) is recommended as the standard approach for tumors of the body and tail of the pancreas. It offers advantages in terms of delayed postoperative gastric emptying, functional recovery time, and length of hospital stay compared to OPD[77,78]. In 2023, the study “minimally invasive vs open distal pancreatectomy for resectable pancreatic cancer” was published in Lancet, demonstrating the non-inferiority of MIDP compared to OPD in terms of resection rates. Equivalent rates of R0 resection, median lymph node harvest, and intraperitoneal recurrence were found. In addition, the median functional recovery time and length of hospital stay were comparable, as were the one- and two-year survival rates[79,80]. According to a European study[81], the annual use of robotic distal pancreatectomy (RDP) increased from 30.5% to 42.6% and was associated with fewer grade intraoperative events compared to laparoscopic distal pancreatectomy, although with a longer operating time. No significant differences were observed in terms of morbidity and hospital/30-day mortality. RPD was associated with fewer conversions in overweight patients, in cases of previous abdominal surgery, and in cases of vascular involvement[81]. Chen et al[82] demonstrated that the RDP group had higher rates of R0 resection, a greater number of lymph nodes removed, and a greater number of vascular resections, indicating that the RDP group underwent more extensive surgery, which could explain the longer hospital stay.
Total pancreatectomy may be essential to surgically extirpate multicentric pancreatic cancer[83]. Minimally invasive total pancreatectomy and open total pancreatectomy have comparable operative and oncological outcomes[83]. Scholten et al[84] reported improved conversion rates for robotic total pancreatectomy at 13.3% vs 42% for laparoscopic total pancreatectomy, with otherwise similar outcomes.
Metastases in PDAC have historically been a contraindication to curative resection. Some studies have reported the technical feasibility and safety of metastasectomy for oligometastatic disease, defined as disease with three or fewer metastatic lesions in the liver, lungs, or both[85-90]. The clinical status of “oligometastasis” is still poorly understood. Its adoption remains controversial, and current evidence is not strong enough to influence guidelines. Metastasectomy is justified when it is safe and offers a benefit in terms of survival or improved quality of life.
The liver is the most common organ for initial metastatic spread or distant recurrence in patients with pancreatic cancer[88] and the median survival has remained poor compared with other sites of metastases[91]. Frigerio et al[92] studied patients with synchronous liver-only metastases (73% had more than two liver metastases). All patients underwent preoperative chemotherapy (63.5% FOLFIRINOX, 36.5% gemcitabine), achieving complete regression of metastatic lesions prior to surgery. In 67.3% of patients, CA19-9 normalization was observed after treatment. With an R0 resection rate of 86.5%, OS from diagnosis was 37.2 months and median DFS after pancreatectomy was 16.5 months. A recurrence occurred in 75% of cases. According to a multivariate analysis, the main factor associated with recurrence was omission of adjuvant therapy. The improvement in OS demonstrates the importance of chemotherapy in helping to select favorable biology[93].
Patients with pulmonary metastases have been shown to have a longer time between pancreatectomy and recurrence and better OS than those with other types of recurrence[94]. Pulmonary metastasectomy has, in recent years, been recognized as a procedure for patients with pancreatic cancer, with reported 5-year survival rates of 31%-70%[95-99].
Peritoneal carcinomatosis has always been considered incurable and treated palliatively with chemotherapy (median OS: 1.5-17 months[100]). The standard regimens are gemcitabine plus nab-paclitaxel (GnP) or FOLFIRINOX, which modestly increase survival compared to gemcitabine alone[101,102]. However, the median survival for metastatic pancreatic cancer ranges from 4.4 to 11.1 months, regardless of the treatment protocol used[103]. Intraperitoneal chemotherapy significantly increases the surgical conversion rate compared to systemic chemotherapy for peritoneal metastases[104-106]. Hy
Patients undergoing cytoreductive surgery (CRS)/HIPEC were compared with patients undergoing systemic therapy alone. A statistically significant difference in median OS, from diagnosis of peritoneal metastasis, was found (19 months for systemic therapy alone and 41 months for CRS/HIPEC). In addition, OS at 1, 2, and 3 years was 81%, 31%, and 8% for systemic therapy alone, compared with 91%, 66%, and 59% for CRS/HIPEC. From the time of surgery, median OS was 26 months, with OS at 1, 2, and 3 years being 76%, 57%, and 39%, respectively. The median progression-free survival (PFS) was 13 months. These results suggest that a selected group of patients may benefit from CRS/HIPEC treatment. However, the procedure is not considered the standard of care for PDAC with peritoneal metastases. Further investigation is needed and is currently underway (No. NCT04858009)[108,109].
In addition to HIPEC, there are also other methods of intraperitoneal chemotherapy, namely normothermic intraperitoneal chemotherapy (NIPEC) and pressurized intraperitoneal aerosol chemotherapy (PIPAC). A study compared conventional systemic therapy and NIPEC with paclitaxel, demonstrating improved survival in the intraperitoneal therapy group (17.9 months vs 10.2 months). Patients who responded to treatment and underwent conversion surgery then had a median survival of 27.4 and 11.3 months, respectively[106].
PIPAC allows for homogeneous and deep penetration of chemotherapy into the peritoneum through the production of aerosols. In a phase II study, a PIPAC approach was used with the administration of cisplatin and doxorubicin, achieving a median survival of 15.6 months[110]. Di Giorgio et al[111] found pathological regression in 50% of cases using PIPAC with oxaliplatin or cisplatin-doxorubicin in patients with peritoneal metastases of pancreatic and biliary origin.
A median survival of 10 months and a 1-year survival rate of 50% were achieved in a phase I study of patients with unresectable peritoneal metastases from various tumors[112,113].
Frassini et al[104] systematically reviewed complications following HIPEC, PIPAC, and NIPEC. The incidence of grade III and IV side effects was 5.5%, 5.1%, and 6.2%, respectively. Intraperitonel chemotherapy therefore has the potential to improve outcomes for pancreatic cancer with peritoneal metastases, however, it can only be applied in the clinical trial setting.
The treatment process varies depending on the stage of the tumor. For resectable pancreatic cancer, surgical resection combined with adjuvant chemotherapy is recommended[114]. The most successful regimens are modified FOLFIRINOX (mFOLFIRINOX) and gemcitabine-capecitabine[114-116]. In 2018, the PRODIGE-24/CCTG PA6 study demonstrated that adjuvant therapy with mFOLFIRINOX significantly improves median DFS (21.6 months vs 12.8 months) and OS (54.4 months vs 35.0 months) compared to gemcitabine alone[114]. In Asia, the S-1 regimen has become the standard of care following the JASPAC 01 study. It is a phase III randomized study conducted in Japan reported that adjuvant chemotherapy with S-1 offered a 5-year OS of 44.1% and a median OS of 46.5 months vs 24.4% and 25.5 months for gemcitabine alone[116]. However, S-1 is not as widely used in America and Europe, mainly because the maximum tolerated dose in Caucasians is lower.
It should be noted that one-third of patients do not receive adjuvant chemotherapy, mainly due to postoperative surgical complications[117]. In patients with resectable pancreatic cancer, randomized trials have not consistently shown benefits with the use of neoadjuvant chemotherapy[118]. However, resectability was previously evaluated only from an anatomical point of view. The 2024 National Comprehensive Cancer Network guidelines, considering activity based classification (A-B-C) approach, also provide for the possibility of treating resectable PDAC with neoadjuvant therapy if high-risk disease features are present (e.g., large primary tumor, regional lymphadenopathy, markedly elevated serum CA19-9, and excessive weight loss)[58,119,120]. In patients with resectable pancreatic cancer, the rate of resection after neoadjuvant therapy is 77%[121]. For patients with BRPC, the standard treatment is neoadjuvant chemotherapy, followed by surgical resection and adjuvant chemotherapy[30,54,58]. There are still no conclusive data regarding the best neoadjuvant chemotherapy regimen. The phase III PREOPANC-2 study[122] is very interesting. It highlights how, in patients with resectable pancreatic cancer and BRPC, FOLFIRINOX in the neoadjuvant setting did not improve OS compared to gemcitabine-based chemoradiotherapy[122].
The resection rate after neoadjuvant therapy for BRPC is 61%[121]. Whether the addition of radiotherapy to che
Patients with LAPC are mainly treated with systemic chemotherapy for 4-6 months, using mFOLFIRINOX or gemcitabine-nab-paclitaxel regimens[54,58]. Trials showed similar OS between the two regimens[126-128]. About 22% of patients initially diagnosed with LAPC undergo resection after induction chemotherapy[121]. Radiotherapy is associated with increased rates of R0 resection and pathological complete response[129-131]. However, few randomized trials have not shown a survival benefit of adding preoperative radiotherapy, as the ALLIANCE A021501[132]. According to a study by Franklin et al[133], after multi-agent neoadjuvant chemotherapy and resection for pancreatic cancer, additional adjuvant chemoradiotherapy vs adjuvant chemotherapy alone is associated with improved survival for patients with lymphatic-vascular invasion + or grade III tumors. A recent meta-analysis reported that for patients with LAPC who are unlikely to receive resection, neoadjuvant radiotherapy seems to improve OS, PFS, DFS, and recurrence-free survival[134].
For patients with advanced or metastatic disease, systemic chemotherapy is the mainstay of treatment. For decades, 5-fluorouracil (5-FU) was the only chemotherapy option for treating pancreatic cancer. Subsequently, gemcitabine was approved as a first-line treatment for pancreatic cancer in 1997 and in 2003 a multi-agent regimen (FOLFIRINOX) was successfully used for metastatic PDAC[107]. FOLFIRINOX improves OS, PFS, and overall response rate (ORR) compared to gemcitabine in patients with metastatic pancreatic cancer and good performance status (ECOG). The PRODIGE 4/ACCORD 11 study[102] reported a median OS of 11.1 months in the FOLFIRINOX-treated group and 6.8 months in the gemcitabine-treated group.
According to the PANOPTIMOX-PRODIGE 35 study, median survival without deterioration in quality of life is better with a regimen based on 4 months of FOLFIRINOX followed by leucovorin plus maintenance treatment with 5-FU[135]. Recently, NALIRIFOX (liposomal irinotecan, 5-FU, oxaliplatin, and leucovorin) has been approved by the Food and Drug Administration (FDA) as a first-line treatment for metastatic PDAC[136]. This approval was based on the results from the phase 3 NAPOLI-3 trial, which demonstrated significant improvements in OS (11.1 months vs 9.2 months) and PFS (7.4 months vs 5.6 months) in comparison with GnP[137]. NALIRIFOX used a lower chemotherapy dosage than FOLFI
After chemotherapy, it is necessary to evaluate the response obtained. In restaging, the main radiological parameter considered is the exclusion of progression by metastatic disease, which is a real challenge due to the difficulty in distinguishing residual tumor tissue from fibrosis on CT[58,140]. An A-B-C approach is therefore necessary in the re-evaluation. To consider surgery in patients with BRPC, the serum CA19-9 concentration must be at least stable, while in patients with LAPC, a substantial decrease after induction chemotherapy is necessary[58]. In patients with CA19-9 not elevated at diagnosis, the biological response is assessed by fluorodeoxyglucose positron emission tomography[141]. As can be inferred from the overall complexity, staging, restaging, and subsequent therapeutic decisions must be followed by a multidisciplinary team.
Mutations in the homologous recombination genes BRCA1, BRCA2, and PALB2 are found in 15%-19% of patients with PDAC. In these cases, platinum-based chemotherapy and poly (adenosine diphosphate-ribose) polymerase inhibitors (PARPi) have been associated with improved PFS and OS. Wattenberg et al[142] reported that PDAC patients with germline BRCA1 and BRCA2 mutations had a higher ORR than those without these mutations (71.4% vs 13.9%) when treated with FOLFIRINOX. A randomized phase II trial comparing cisplatin-gemcitabine with and without veliparib in patients with BRCA and PALB2 mutations reported ORRs of 74.1% and 65.3%, respectively[143].
In the POLO trial[144], patients with metastatic PDAC with BRCA1/BRCA2 mutations and stable disease or who responded to treatment after 4 months of platinum-based chemotherapy were randomized to receive olaparib or placebo.
The group receiving olaparib showed longer PFS (7.4 months vs 3.8 months). Following this study, olaparib was approved by the European Medicines Agency and FDA as maintenance therapy in patients with metastatic PDAC with BRCA1/BRCA2 mutations whose disease had not progressed after at least 4 months of first-line platinum-based therapy.
Subsequently, the RUCAPANC2 study[145] demonstrated that rucaparib can improve median OS, reaching 23.5 months. Reiss et al[145] identified BRCA or PALB2 reversion variants that restore the function of the corresponding proteins in patients whose disease had progressed during maintenance therapy with PARPi. Patients with these variants developed rapid resistance to PARPi and had worse outcomes than those without reversions, with PFS of 3.7 months vs 12.5 months and OS of 6.2 months vs 23.0 months. Future research should focus on developing treatment options for pancreatic cancer patients who are resistant to PARPi and platinum-based therapies[146].
New studies are evaluating the use of olaparib as adjuvant therapy (APOLLO/EA2192, No. NCT04858334), melphalan (No. NCT04150042) and pidnarulex (No. NCT04890613).
In PDAC, 90%-92% of cases have a driver mutation in the KRAS oncogene, which classifies PDAC genomically into two subtypes, KRAS mutant (KRASmut) and KRASwt PDAC[8,39]. In pancreatic cancer, the most common KRAS mutations are KRAS G12D (40%), followed by G12V (29%), G12R (15%), and less commonly G12C (approximately 1%)[147]. Cur
Adagrasib and sotorasib are small-molecules, covalent inhibitors that irreversibly and selectively bind KRAS G12C, trapping it in its inactive guanosine diphosphate-bound state. Adagrasib and sotorasib demonstrated good results in patients with KRAS G12C mutations in the CodeBreak 100 and KRYSTAL-1 trials, respectively[75,147,148]. Additional KRAS G12C inhibitors in clinical trials include JDQ443 (No. NCT04699188), JAB-21822 (No. NCT05002270), D-1553 (No. NCT04585035), GDC-6036 (No. NCT04449874), LY3537982 (No. NCT04956640), and BI-1823911 (No. NCT04973163).
MRTX-1133, a novel inhibitor targeting KRAS G12D mutations, is in phase I clinical trials and has shown promising preclinical data[149]. A phase I trial (No. NCT05737706) is currently exploring the use of MRTX-1133 in KRAS G12D advanced solid tumors[150]. Another promising molecule is RMC-6236, which targets multiple RAS variants[151]. The phase I RMC-6236-001 study enrolled patients with advanced tumors harboring mutations at codon 12 of KRAS (KRAS G12X), excluding KRAS G12C, after progression on at least one standard therapy option[152]. The most recent updated analysis showed that the 14 + week disease control rate in KRAS G12X mutated PDAC was 87% with a median PFS of 8.1 months. Patients with metastatic PDAC and KRAS G12X mutation in the second-line setting, achieved a median PFS of 8.5 months and a median OS of 14.5 months. The response rate for patients harboring KRAS G12X mutations was 29% in the second-line group and 22% in the third line and beyond. Lower outcomes were reported in patients harboring any RAS mutation. These promising results have led to the initiation of RASolute 302 (No. NCT06625320), a phase 3 ran
New and very interesting clinical trials are underway on indirect targets to block KRAS. A phase I trial in patients with advanced KRAS-mutated cancers is evaluating the safety and efficacy of BI1701963, an inhibitor of the nucleotide exchange factor SOS1, alone and in combination with trametinib, a MAPK pathway inhibitor (No. NCT04111458)[155,156]. Multiple studies (No. NCT04418661, No. NCT04330664, and No. NCT03634982) are investigating combinations to block Src homology 2-containing protein tyrosine phosphatase 2, a tyrosine phosphatase necessary for KRAS activation, and ASP3082 (No. NCT05382559), capable of binding ubiquitin ligase E3 and KRAS G12D, thus allowing the ubiquitination of the latter and its degradation by the proteasome[157-159]. Lastly, RMC-7977 is a highly selective inhibitor of the active guanosine triphosphate-bound forms of KRAS, HRAS, and NRAS, with affinity for both mutant and wild-type variants. It has shown promising results in preclinical models[160,161].
KRASwt PDAC can harbor other mutations, including rare gene fusions involving FGFR2, RAF, ALK, RET, MET, NTRK1, ROS1, ERBB4, NRG1, and FGFR3, among others. Although these genetic fusions are uncommon, they can offer an opportunity for personalized therapeutic approaches. Zenocutuzumab recently received FDA approval for advanced or metastatic NRG1 fusion-positive pancreatic cancer[75,162]. BRAF V600E is rare, present in approximately 3% of PDAC cases. The combination of dabrafenib and trametinib, which targets BRAF V600E mutations, was investigated in the NCI-MATCH trial and the ROAR basket trial, demonstrating efficacy and a manageable toxicity profile[163]. In 2022, the FDA granted accelerated approval for the use of this combination in adult and pediatric patients (≥ 6 years) with unresectable or metastatic BRAF V600E-positive solid tumors who had progressed after prior therapy and lacked other suitable options. Additionally, selpercatinib, a kinase inhibitor targeting RET fusions, has received FDA accelerated approval based on promising results in RET fusion-positive solid tumors, non-small cell lung cancer, medullary thyroid cancer, and thyroid cancer[164,165]. A phase II trial (No. NCT04390243) is currently investigating the combination of encorafenib, a BRAF inhibitor, and binimetinib, a mitogen-activated protein kinase kinase inhibitor, in patients with pretreated metastatic PDAC with somatic BRAF V600E mutations. Furthermore, in 2024, the FDA granted accelerated approval to fam-trastuzumab deruxtecan for adult patients with unresectable or metastatic human epidermal growth factor receptor 2 (HER2)-positive (immunohistochemistry 3 +) solid tumors, based on data from the DESTINY-PanTumor2 trial[166]. Larotrectinib and entrectinib are recommended as first-line treatment options for locally advanced or metastatic pancreatic cancer with NTRK fusions[167,168]. Repotrectinib, a newer NTRK inhibitor, received accelerated FDA approval in 2023, showing promising results in the TRIDENT trial[75,169,170].
Uncontrolled cell division in pancreatic cancer causes high levels of replicative stress and stimulates activation of the ataxia telangiectasia and Rad3-related serine/threonine kinase-checkpoint kinase 1 (CHK1)-WEE1 pathway[170,171]. The inhibition of this pathway is currently under investigation. The use of a single agent has shown poor efficacy and significant myelosuppression[172]. Better results were obtained by combining the SRA737 CHK1 inhibitor with gemcitabine. In this case, although the ORR was 10.8%, the therapy was well tolerated with few side effects[173]. In preclinical models, PARPi-resistant pancreatic cancer cells have been shown to be highly sensitive to the WEE1 inhibitor adavosertib[174]. In a phase I study, the combination of radiotherapy with gemcitabine and adavosertib in patients with advanced pancreatic cancer demonstrated a median PFS of 9.4 months and a median OS of 21.7 months[175]. An interesting target for PDAC is FGFR2 fusions. Stein et al[176], in 2024, showed a case series of four FGFR2 fusion-positive metastatic PDAC patients who achieved durable responses with fibroblast growth factor receptor inhibitors. REFOCUS is a phase I/II trial evaluating the highly selective FGFR2 inhibitor, RLY-4008, in patients with intrahepatic cholangiocarcinoma and other advanced solid tumors (No. NCT04526106)[177]. Another target currently being explored is methylthioadenosine phosphorylase (MTAP) or CDKN2A-deleted cancers, including pancreatic cancer. AMG 193, a PRMT5 inhibitor targeting MTAP or CDKN2A-deleted tumors, showed promising antitumor activity and a favorable safety profile in a phase I study involving 80 patients with MTAP-deleted solid tumors[178]. AMG 193 is currently being evaluated in clinical trials in combination with chemotherapy in patients with MTAP-deleted PDAC (No. NCT06360354).
Microsatellite instability and deficiency mismatch repair proteins (MSI-H/dMMR) are associated with increased immunogenicity and significant responsiveness to immune checkpoint inhibitors (ICI)[179,180]. Based on positive outcomes from clinical trials involving 504 patients across different cancer types (KEYNOTE-158, KEYNOTE-164, KEYNOTE-051), the FDA granted full approval for pembrolizumab monotherapy for patients with unresectable MSI-H/dMMR solid tumors in second-line settings and beyond[180-182]. However, MSI-H/dMMR occurs in only 1% of PDAC cases, and its use remains debated. Results from the KEYNOTE-158 trial indicated lower responses (18.2%) in PDAC patients treated with pembrolizumab compared to other non-colorectal cancers (31%-48.5%)[180]. Real-world data from retrospective studies conducted at the Mayo Clinic and from the Association des Gastro-Enterologue Oncologues European cohort demonstrated better outcomes[183,184] and support the use of ICIs in this patient population, especially if MSI-H/dMMR is discovered.
The junctional protein claudin 18.2 is overexpressed in gastric and pancreatic tumors. Zolbetuximab, a monoclonal antibody against claudin 18.2, in combination with chemotherapy has shown promising results in terms of PFS and OS in both the phase III SPOTLIGHT and GLOW studies in patients with claudin 18.2-positive gastric and gastroesophageal junction tumors (No. NCT03504397 and No. NCT03653507). A randomized study is currently evaluating the combination of GnP with or without zolbetuximab in patients with claudin 18.2-positive metastatic pancreatic cancer (No. NCT03816163). A phase I study (No. NCT05482893) is evaluating PT886, a bispecific antibody that targets claudin 18.2 and cluster of differentiation (CD) 47, in patients with unresectable or metastatic gastric tumors, gastroesophageal junction tumors, and pancreatic cancer[185].
High levels of αvβ5 integrins are expressed in human PDAC but not in healthy tissues. A non-randomized study evaluated the use of certepetide (CEND-1), a peptide that specifically targets αv integrins, administered in combination with GnP in 31 patients with previously untreated metastatic PDAC[186,187]. ORR was 59%, PFS was 9.7 months, and median OS was 13.2 months[186]. Based on these results, a randomized, double-blinded phase II trial is currently enrolling patients (No. NCT05042128) in Australia and New Zealand. In the United States, CENDIFOX (No. NCT05121038) is an ongoing phase Ib-IIa trial of CEND-1 in combination with neoadjuvant FOLFIRINOX in pancreatic, colorectal, and appendiceal cancers[188]. The target therapy for PDAC has been summarized in Table 1.
| Category | Details |
| Homologous recombination deficiency | Prevalence: Mutations in homologous recombination genes (BRCA1, BRCA2, PALB2) are found in 15%-19% of patients with PDAC. Therapeutic response: Patients with HRD show improved PFS and OS after platinum-based chemotherapy and PARPi. Retrospective analysis: In a study of patients treated with FOLFIRINOX, the ORR was significantly higher in BRCA1/BRCA2 mutated patients (71.4%) compared to non-mutated patients (13.9%). Platinum-based therapy response: A separate retrospective study reported an ORR of 58% in patients with germline BRCA and PALB2 mutations compared to 21% in the control group |
| PARPi trials | POLO trial: This trial evaluated olaparib as maintenance therapy for patients with germline BRCA1/BRCA2 mutations who had stable or responding disease after 4 months of platinum-based chemotherapy. The study found a significant increase in PFS (7.4 months for olaparib vs 3.8 months for placebo), although there was no significant difference in OS (19 months for olaparib vs 19.2 months for placebo). FDA approval: Olaparib has been approved as maintenance therapy for platinum-sensitive metastatic PDAC patients with BRCA1/BRCA2 mutations whose disease has not progressed after at least 16 weeks of first-line platinum-based treatment. RUCAPANC2 trial: This phase II single-arm trial demonstrated that rucaparib, administered as maintenance therapy in patients with advanced pancreatic cancer harboring germline or somatic BRCA or PALB2 mutations, achieved a median OS of 23.5 months. Reversion variants: In a study by Reiss et al[145], 16.6% of patients whose disease progressed on maintenance PARPi developed BRCA or PALB2 reversion variants, which restored the function of the BRCA or PALB2 proteins. Patients with these reversion variants experienced rapid resistance to PARPi (PFS of 3.7 months vs 12.5 months; P = 0.001) and had significantly poorer OS (6.2 months vs 23.0 months; P < 0.0001) |
| KRAS mutations | Prevalence: KRAS mutations are present in 90%-92% of PDAC cases, classifying them into KRASmut and KRASwt subtypes. Common mutations: The most frequent KRAS mutations include G12D (40%), G12V (29%), G12R (15%), and G12C (approximately 1%). Therapeutic challenge: Currently, there are no FDA-approved therapies specifically targeting KRASmut PDAC, making it a significant area of unmet medical need |
| KRAS G12C inhibitors | Inhibitors: Adagrasib and sotorasib are small-molecule covalent inhibitors that irreversibly bind to KRAS G12C, trapping it in its inactive guanosine diphosphate-bound state. Clinical trials: Both inhibitors have shown promising results in patients with KRAS G12C mutations in the CodeBreak 100 and KRYSTAL-1 trials, demonstrating efficacy and manageable safety profiles. Other inhibitors: Additional KRAS G12C inhibitors currently in clinical trials include JDQ443 (No. NCT04699188), JAB-21822 (No. NCT05002270), D-1553 (No. NCT04585035), GDC-6036 (No. NCT04449874), LY3537982 (No. NCT04956640), and BI-1823911 (No. NCT04973163) |
| KRAS G12D inhibitors | MRTX-1133: A novel inhibitor specifically targeting KRAS G12D mutations, currently in phase I clinical trials, has shown promising preclinical data. RMC-6236: This inhibitor targets multiple RAS variants and is undergoing a phase I study. The most recent analysis indicated a 14 + week disease control rate of 87% in patients with KRAS G12X mutations, with a median PFS of 8.1 months. RASolute 302 trial: A phase 3 randomized trial evaluating the efficacy of RMC-6236 in second-line metastatic PDAC, regardless of RAS status, is currently ongoing |
| KRAS wild-type PDAC | Genetic fusions: KRASwt PDAC can harbor rare gene fusions involving FGFR2, RAF, ALK, RET, MET, NTRK1, ROS1, ERBB4, NRG1, and FGFR3, which may provide opportunities for personalized therapeutic approaches. FDA approvals: Zenocutuzumab has received FDA approval for advanced or metastatic NRG1 fusion-positive pancreatic cancer. The combination of dabrafenib and trametinib, targeting BRAF V600E mutations, was investigated in the NCI-MATCH trial and the ROAR basket trial, demonstrating efficacy and a manageable toxicity profile. Ongoing trials: A phase II trial (No. NCT04390243) is currently investigating the combination of encorafenib (a BRAF inhibitor) and binimetinib (a MEK inhibitor) in patients with pretreated metastatic PDAC with somatic BRAF V600E mutations |
| Microsatellite instability | Prevalence: MSI-H/dMMR occurs in only 1% of PDAC cases, which limits the applicability of immunotherapy. Therapeutic response: Pembrolizumab has been granted full FDA approval for unresectable MSI-H/dMMR solid tumors based on positive outcomes from clinical trials (KEYNOTE-158, KEYNOTE-164, KEYNOTE-051). However, response rates in PDAC patients treated with pembrolizumab were lower (18.2%) compared to other non-colorectal cancers (31%-48.5%). Real-world data: Retrospective studies from the Mayo Clinic and the AGEO European cohort suggest better outcomes for MSI-H/dMMR PDAC patients, supporting the use of immune checkpoint inhibitors |
Vaccines and miRNA therapies can target KRAS. The AMPLIFY-201 study (No. NCT04853017) is evaluating the adjuvant regimen ELI-002, a therapeutic vaccine targeting KRAS-mutated tumors, for patients with KRAS G12D, G12R PDAC and colorectal tumors who are in a state of minimal residual disease with a high risk of recurrence[189]. Reductions in CEA and CA19-9 were observed in 79% of patients, while clearance of minimal residual disease was observed in 21%. Mul
Very interesting phase I studies are No. NCT04117087 and No. NCT04161755. The first is currently evaluating a vac
A phase I/II study reported an ORR of 44% and a disease control rate of 94% with the combination of intratumoral injections of LOAd703, an oncolytic adenovirus, and standard chemotherapy with nab-paclitaxel/gemcitabine in patients with unresectable or metastatic PDAC (No. NCT02705196)[194].
The combination of the reovirus pelareorep and pembrolizumab, on the other hand, showed modest efficacy with a clinical benefit rate of 42%. However, led to significant immunological changes in CD8+ T cells and CD4+ regulatory T cells during treatment in patients who responded to therapy (No. NCT03723915)[195].
Finally, a clinical trial will evaluate the efficacy of talimogene laherparepvec, administered endoscopically, for the treatment of locally advanced or metastatic pancreatic cancer refractory to at least one chemotherapy regimen (No. NCT03086642).
Chimeric antigen receptor (CAR) T-cell therapy involves the use of receptors expressed on genetically modified T cells, which are able to recognize specific surface antigens on tumor cells and thus kill them[196]. It is a promising approach, especially against tumors that are resistant to standard therapies. The antigens currently being studied for CAR-T therapy in pancreatic cancer are mesothelin, CEA, MUC1, epidermal growth factor receptor (EGFR), CD133, and claudin 18.2[172,197].
Several phase I clinical trials have reported partial responses in a small number of patients[198-200]. The phase I study (No. NCT02159716) examined lentivirus-transduced CAR-T cells targeting mesothelin in patients with metastatic pancreatic cancer. Of the five patients, three showed no response and two had disease stabilization for only two to three months. Further disappointing results were obtained in a phase I study (No. NCT01869166) that evaluated EGFR CAR-T cells after a chemotherapy regimen based on nab-paclitaxel plus cyclophosphamide. The median PFS and OS obtained were 3 and 4.9 months, respectively[198].
Research is currently underway with fully human anti-mesothelin CAR (No. NCT03054298 and No. NCT03323944). Several phase I studies[199-203] have evaluated the use of autologous CAR-T cells against claudin 18.2. They reported poor results in terms of efficacy, with only a small percentage of patients achieving partial response or disease stabilization, and significant hematological toxicity[204].
Apparently more promising results have been obtained with anti-MUC1 CAR-T cells, which have demonstrated ef
Two studies (No. NCT05239143 and No. NCT04025216[206-208]), were conducted to evaluate the clinical application of anti-MUC1 CAR-T cells. Numerous clinical trials have been registered for the use of EPCAM-specific CAR-T cells in individuals with pancreatic cancer (No. NCT04151186 and No. NCT03013712) and ROBO1- and MUC1-specific CAR-natural killer cells (No. NCT03941457 and No. NCT02839954)[208].
In general, immunotherapy has revolutionized the field of oncology. However, in the case of pancreatic cancer, studies (preclinical and clinical) have highlighted the role of the tumor microenvironment (TME) as an obstacle to CAR-T cell therapy. Other limitations that hinder the effectiveness of CAR-T cells in pancreatic cancer are heterogeneous antigen expression and cell-mediated toxicities.
The latest development involves T cells engineered to express T cell receptors (TCRs) capable of recognizing tumor antigens[209]. Chiorean et al[210] have registered a study of the safety and efficacy of MSLN-specific autologous T cells in patients with stage IV pancreatic cancer (No. NCT04809766). MSLN-specific autologous TCR T cells are used in com
Recent discoveries in the field of immunotherapy involve the CD39/CD73 axis and bispecific antibodies (BsAbs). CD39 and CD73 are ectonucleotidases that allow the accumulation of extracellular adenosine, which suppresses the immune system in both innate and adaptive responses. The anti-CD73/CD39 antibodies currently being investigated are ole
BsAbs are specialized antibodies designed to act effectively on two specific antigens simultaneously[213]. BsAbs have been developed for pancreatic cancer treatment, with examples such as anti-EGFR × HER2[214], anti-CD3 × CEA[215], MCLA-128 (anti-HER2 × HER3 BsAb; zenocutuzumab)[216], anti-CD3 × EGFR BsAb[217], anti-CD3 (Vγ9TCR) × HER2/Neu[218], XmAb22841 [anti-LAG3 × cytotoxic T-lymphocyte-associated protein 4 (CTLA-4); No. NCT03849469], XmAb
Based on the poor results with single-agent immunotherapy or target therapy, several researchers are recently trying to achieve increased outcomes with combinations of drugs with different mechanisms.
PDAC with biallelic loss of BRCA1 and BRCA2 genes has greater sensitivity to ICIs[221] than wild-type tumors because it has a higher mutation burden. A retrospective study evaluated patients with platinum-refractory metastatic pancreatic or biliary carcinoma with mutations in homologous recombination genes who were treated with ipilimumab and nivo
In a recent phase Ib/II trial (PARPVAX trial), patients who had not progressed on platinum-based first-line therapy were randomized to receive niraparib plus nivolumab or niraparib plus ipilimumab as maintenance therapy[224]. The results were promising, with a 6-month PFS of 20.6% in the niraparib plus nivolumab group and 59.6% in the niraparib plus ipilimumab group. Based on these encouraging data, several studies are in progress. New results from the POLAR study on metastatic PDAC were presented at the European Society for Medical Oncology 2024 congress[225]. Patients were treated with pembrolizumab plus olaparib and were enrolled in three different cohorts: (1) Homologous recombination deficiency (HRD) patients with BRCA1/BRCA2 or PALB2 mutations; (2) Patients with non-core HRD mutations such as ATM; and (3) Patients with no HRD but with a high response to platinum therapy. Cohort A showed the most promising results, with 64% of patients without PFS at 6 months and a disease control rate of 90%. Based on the POLO study[170] and the immunostimulatory capacity of PARPs[226-228], the SWOG0G2001 study (No. NCT04548752) is evaluating the combination of olaparib and pembrolizumab vs olaparib alone with the primary objective of increasing median PFS.
Clinical trials are also examining PARPis combined with FOLFIRI (No. NCT02498613). These combinations may increase the sensitivity to PARPis for a larger population than that with BRCA mutations. These approaches are highly innovative and still need to be evaluated in well-designed clinical trials.
The main obstacle to old and new therapies is the TME. The TME constitutes the majority of the tumor complex (approximately 70% of the total volume), while only a small percentage is represented by PDAC neoplastic cells. The pancreatic TME consists of numerous populations of fibroblasts, a dense extracellular matrix, and immune cells with suppressive function. The result is a desmoplastic stroma that compresses vascular structures and creates a hypoxic environment, thereby affecting the pharmacokinetics and pharmacodynamics of therapy. The dense component also prevents immune system cells from reaching the target site. What we are therefore faced with is a chemoresistant and immunoresistant tumor[229]. Targeting fibrosis in pancreatic cancer is crucial due to its significant impact on treatment efficacy. For this reason, several studies are evaluating the use of standard treatments with new agents against stromal components, such as pamrevlumab, a new monoclonal antibody that binds to connective tissue growth factor (No. NCT04229004), or a pegylated recombinant hyaluronidase (No. NCT02715804). Other interesting studies are NCT03727880 and STARPAC[230]. The first evaluates the use of pembrolizumab with defactinib, a focal adhesion kinase inhibitor, as neoadjuvant and sequential adjuvant therapy in patients with high-risk resectable PDAC[172]. The second is a multicenter, randomized, controlled clinical trial in LAPC that tests whether a combination of standard gemcitabine and nab-paclitaxel che
Recent advances in AI have shown encouraging results in: (1) Detecting and segmenting the pancreas and pancreatic lesions; (2) Classifying lesions as benign or malignant; and (3) Developing predictive models for early diagnosis[231].
Cao et al[232] developed an approach to detect and classify pancreatic lesions with high accuracy using non-contrast CT with AI (like PANDA). PANDA was trained on a dataset of over 3000 patients and achieves an AUC of 0.986-0.996 for lesion detection in a multicenter validation. It outperforms the average performance of radiologists by 34% in terms of sensitivity and 6% in terms of specificity for PDAC identification and achieves a sensitivity of 93% and a specificity of 99% for lesion detection in a validation involving 20530 consecutive patients. In particular, PANDA shows non-inferiority compared to radiological reports using contrast-enhanced CT and could potentially serve as a new tool for large-scale screening for pancreatic cancer.
Equally good results were obtained in studies conducted in China[233,234]. Si et al[233] developed a deep learning model with an average accuracy for all tumor types of 82.7%, and independent accuracies of identifying intraductal papillary mucinous neoplasm and PDAC of 100% and 87.6%, respectively.
With AI, the diagnostic efficiency of EUS images has also been significantly improved, especially in the diagnosis of autoimmune pancreatitis[235,236]. A case in point is the model developed by Marya et al[235], trained using static images and videos from EUS examinations on patients. The model achieves high levels of sensitivity and specificity in differentiating autoimmune pancreatitis from PDAC, from normal pancreas, and from chronic pancreatitis.
In addition to imaging, AI is exploring the integration of clinical data to create predictive models that can identify individuals at high risk of developing PDAC, enabling more targeted screening programs and, potentially, the implementation of noninvasive tests for early diagnosis. Placido et al[237] analyzed clinical data from 6 million patients, including 24000 with PDAC in the Danish National Patient Registry and 3 million patients, with 3900 cases of PDAC, in the United States Department of Veterans Affairs databases.
The best-performing model was able to predict the onset of pancreatic cancer within 36 months of initial diagnosis based on data extracted from electronic medical records with an AUROC curve of 0.879.
AI is also optimizing therapeutic planning. AI can analyze the genetic and molecular profile of the tumor, combining it with the clinical characteristics of the patient, to predict the response to different chemotherapies or targeted therapies. This data-driven approach makes it possible to select the most effective treatment regimen for the individual patient, avoiding ineffective treatments and reducing toxicity[238]. For example, AI algorithms can identify specific mutations and suggest approved or clinical trial drugs that target them. AI is accelerating the discovery of new therapeutic targets. By analyzing vast databases of tumor genomes, proteomes, and clinical data, AI can identify novel signaling pathways involved in PDAC growth and progression, suggesting potential drugs that block them[239]. This includes finding new strategies to overcome drug resistance, a persistent problem in PDAC treatment. AI is not only improving clinical pra
Although AI models are making progress and have had noteworthy practical triumphs, there are significant obstacles to their application in clinical practice[241]. First, statistical limitations such as the retrospective study model, inability outside the trained domain, unintentional confounding, use of selectively chosen images for algorithm training reflecting possible selection bias, false positive (FP) and false negative detections. A major disadvantage of addressing FP detections is that there is no obvious cause for FP detections to improve model capabilities[242]. The complexity of AI-based ap
Therefore, it is necessary to design and define uniform processes for data collection, processing, storage, replication, and analysis. In addition, experts from institutions located in different geographical areas should collaborate to establish benchmarks for AI-based diagnosis. Finally, there are ethical and legal aspects. To date, AI-assisted image analysis and machine learning neural networks in disease diagnosis and prediction have only provided an “opinion”, as the responsible physician retains complete authority over the decisions to be made. Scientists and engineers are making tremendous progress in the use of AI in their fields, achieving task autonomy or conditional autonomy and, ultimately, complete automation. The responsible party remains unclear if an algorithm makes a mistake with potentially fatal consequences.
The most recent data registry reports an overall 5-year survival rate for pancreatic cancer of 12%, up from 6% for patients diagnosed in 2004. However, for metastatic disease, the 5-year survival rate has only increased from 2% to 3%, suggesting minimal progress, while for localized disease it has increased from 24% to 46%. It is important to note that only 14% of patients are diagnosed at a resectable stage, meaning that the survival rate for most patients remains unchanged. Survival rates for metastatic disease at 4, 3, and 2 years have not increased, but 1-year survival has increased from 14% to 22% (2004-2019). These trends suggest progress in short-term outcomes[244]. The resilience and ability of PDAC to withstand significant stresses underlies the nearly uniform fate of ultimate progression. The microenvironment surrounding PDAC, the main proponent of the tumor’s intrinsic therapeutic resistance, contains potentially crucial information about treatment vulnerabilities[245]. In summary, 2025 does not mark the discovery of a single solution for pancreatic cancer. Rather, we are in an era of incremental but fundamental advances that are gradually shifting the needle in favor of patients. The integration of a multimodal approach that embraces personalized screening, advanced diagnostics, innovative surgery, and increasingly targeted therapies will be the key to transforming the prognosis of this devastating disease. There is still a long way to go, but the foundation for a more promising future has been laid.
| 1. | Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229-263. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 5690] [Cited by in RCA: 9854] [Article Influence: 9854.0] [Reference Citation Analysis (3)] |
| 2. | Stoffel EM, Brand RE, Goggins M. Pancreatic Cancer: Changing Epidemiology and New Approaches to Risk Assessment, Early Detection, and Prevention. Gastroenterology. 2023;164:752-765. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 254] [Reference Citation Analysis (1)] |
| 3. | Blackford AL, Canto MI, Dbouk M, Hruban RH, Katona BW, Chak A, Brand RE, Syngal S, Farrell J, Kastrinos F, Stoffel EM, Rustgi A, Klein AP, Kamel I, Fishman EK, He J, Burkhart R, Shin EJ, Lennon AM, Goggins M. Pancreatic Cancer Surveillance and Survival of High-Risk Individuals. JAMA Oncol. 2024;10:1087-1096. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 4] [Cited by in RCA: 54] [Article Influence: 54.0] [Reference Citation Analysis (0)] |
| 4. | Chandana SR, Woods LM, Maxwell F, Gandolfo R, Bekaii-Saab T. Corrigendum to "Risk factors for early-onset pancreatic ductal adenocarcinoma: A systematic literature review" [Eur J Cancer 198 (2024) 113471]. Eur J Cancer. 2024;201:113941. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
| 5. | Park W, Chawla A, O'Reilly EM. Pancreatic Cancer: A Review. JAMA. 2021;326:851-862. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 798] [Cited by in RCA: 1184] [Article Influence: 296.0] [Reference Citation Analysis (0)] |
| 6. | Blackford AL, Canto MI, Klein AP, Hruban RH, Goggins M. Recent Trends in the Incidence and Survival of Stage 1A Pancreatic Cancer: A Surveillance, Epidemiology, and End Results Analysis. J Natl Cancer Inst. 2020;112:1162-1169. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 60] [Cited by in RCA: 167] [Article Influence: 41.8] [Reference Citation Analysis (0)] |
| 7. | Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209-249. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 75126] [Cited by in RCA: 66304] [Article Influence: 16576.0] [Reference Citation Analysis (183)] |
| 8. | Liew SZH, Ng KW, Ishak NDB, Lee SY, Zhang Z, Chiang J, Ngeow JYY. Geographical, ethnic, and genetic differences in pancreatic cancer predisposition. Chin Clin Oncol. 2023;12:27. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 11] [Reference Citation Analysis (0)] |
| 9. | Abboud Y, Samaan JS, Oh J, Jiang Y, Randhawa N, Lew D, Ghaith J, Pala P, Leyson C, Watson R, Liu Q, Park K, Paski S, Osipov A, Larson BK, Hendifar A, Atkins K, Nissen NN, Li D, Pandol SJ, Lo SK, Gaddam S. Increasing Pancreatic Cancer Incidence in Young Women in the United States: A Population-Based Time-Trend Analysis, 2001-2018. Gastroenterology. 2023;164:978-989.e6. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 29] [Cited by in RCA: 49] [Article Influence: 24.5] [Reference Citation Analysis (0)] |
| 10. | Cronin KA, Scott S, Firth AU, Sung H, Henley SJ, Sherman RL, Siegel RL, Anderson RN, Kohler BA, Benard VB, Negoita S, Wiggins C, Cance WG, Jemal A. Annual report to the nation on the status of cancer, part 1: National cancer statistics. Cancer. 2022;128:4251-4284. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 5] [Cited by in RCA: 346] [Article Influence: 115.3] [Reference Citation Analysis (0)] |
| 11. | Sung H, Siegel RL, Rosenberg PS, Jemal A. Emerging cancer trends among young adults in the USA: analysis of a population-based cancer registry. Lancet Public Health. 2019;4:e137-e147. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 227] [Cited by in RCA: 403] [Article Influence: 67.2] [Reference Citation Analysis (0)] |
| 12. | Gaddam S, Abboud Y, Oh J, Samaan JS, Nissen NN, Lu SC, Lo SK. Incidence of Pancreatic Cancer by Age and Sex in the US, 2000-2018. JAMA. 2021;326:2075-2077. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 34] [Cited by in RCA: 91] [Article Influence: 22.8] [Reference Citation Analysis (0)] |
| 13. | Macarulla T, Hendifar AE, Li CP, Reni M, Riess H, Tempero MA, Dueck AC, Botteman MF, Deshpande CG, Lucas EJ, Oh DY. Landscape of Health-Related Quality of Life in Patients With Early-Stage Pancreatic Cancer Receiving Adjuvant or Neoadjuvant Chemotherapy: A Systematic Literature Review. Pancreas. 2020;49:393-407. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 12] [Cited by in RCA: 20] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
| 14. | Danpanichkul P, Suparan K, Jaroenlapnopparat A, Polpichai N, Fangsaard P, Detboon A, Moolkaew P, Sripusanapan A, Srisurapanont K, Kanjanakot Y, Duangsonk K, Wallace MB, Wijarnpreecha K. The Global Burden of Early-Onset Pancreatic Cancer and Its Risk Factors: A Perspective From Global Burden of Disease Study 2019. Pancreas. 2024;53:e434-e444. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 12] [Cited by in RCA: 11] [Article Influence: 11.0] [Reference Citation Analysis (0)] |
| 15. | Grigorescu RR, Husar-Sburlan IA, Gheorghe C. Pancreatic Cancer: A Review of Risk Factors. Life (Basel). 2024;14:980. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 7] [Reference Citation Analysis (0)] |
| 16. | Pereira SP, Oldfield L, Ney A, Hart PA, Keane MG, Pandol SJ, Li D, Greenhalf W, Jeon CY, Koay EJ, Almario CV, Halloran C, Lennon AM, Costello E. Early detection of pancreatic cancer. Lancet Gastroenterol Hepatol. 2020;5:698-710. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 254] [Cited by in RCA: 317] [Article Influence: 63.4] [Reference Citation Analysis (1)] |
| 17. | Dankner R, Boffetta P, Balicer RD, Boker LK, Sadeh M, Berlin A, Olmer L, Goldfracht M, Freedman LS. Time-Dependent Risk of Cancer After a Diabetes Diagnosis in a Cohort of 2.3 Million Adults. Am J Epidemiol. 2016;183:1098-1106. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 78] [Cited by in RCA: 98] [Article Influence: 10.9] [Reference Citation Analysis (0)] |
| 18. | Jain A, Keesari PR, Pulakurthi YS, Katamreddy R, Dhar M, Desai R. Pancreatic Cancer Risk in Prediabetes: A Systematic Meta-analysis Approach. Pancreas. 2025;54:e51-e56. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 5] [Reference Citation Analysis (0)] |
| 19. | Sapoor S, Nageh M, Shalma NM, Sharaf R, Haroun N, Salama E, Pratama Umar T, Sharma S, Sayad R. Bidirectional relationship between pancreatic cancer and diabetes mellitus: a comprehensive literature review. Ann Med Surg (Lond). 2024;86:3522-3529. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 7] [Reference Citation Analysis (0)] |
| 20. | Yu W, Zhou D, Meng F, Wang J, Wang B, Qiang J, Shen L, Wang M, Fang H. The global, regional burden of pancreatic cancer and its attributable risk factors from 1990 to 2021. BMC Cancer. 2025;25:186. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 5] [Article Influence: 5.0] [Reference Citation Analysis (0)] |
| 21. | Mellenthin C, Balaban VD, Dugic A, Cullati S. Risk Factors for Pancreatic Cancer in Patients with New-Onset Diabetes: A Systematic Review and Meta-Analysis. Cancers (Basel). 2022;14:4684. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 19] [Reference Citation Analysis (0)] |
| 22. | White MJ, Sheka AC, LaRocca CJ, Irey RL, Ma S, Wirth KM, Benner A, Denbo JW, Jensen EH, Ankeny JS, Ikramuddin S, Tuttle TM, Hui JYC, Marmor S. The association of new-onset diabetes with subsequent diagnosis of pancreatic cancer-novel use of a large administrative database. J Public Health (Oxf). 2023;45:e266-e274. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 5] [Reference Citation Analysis (0)] |
| 23. | Deng Z, Gu Y, Hou X, Zhang L, Bao Y, Hu C, Jia W. Association between uric acid, cancer incidence and mortality in patients with type 2 diabetes: Shanghai diabetes registry study. Diabetes Metab Res Rev. 2016;32:325-332. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 13] [Cited by in RCA: 23] [Article Influence: 2.6] [Reference Citation Analysis (0)] |
| 24. | Westermann R, Zobbe K, Cordtz R, Haugaard JH, Dreyer L. Increased cancer risk in patients with cutaneous lupus erythematosus and systemic lupus erythematosus compared with the general population: A Danish nationwide cohort study. Lupus. 2021;30:752-761. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3] [Cited by in RCA: 23] [Article Influence: 5.8] [Reference Citation Analysis (0)] |
| 25. | Peeri NC, Landicino MV, Saldia CA, Kurtz RC, Rolston VS, Du M. Association Between Polycystic Ovary Syndrome and Risk of Pancreatic Cancer. JAMA Oncol. 2022;8:1845-1847. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 5] [Cited by in RCA: 11] [Article Influence: 3.7] [Reference Citation Analysis (0)] |
| 26. | Sun M, Lin JA, Chang CL, Wu SY, Zhang J. Association between long-term opioid use and cancer risk in patients with chronic pain: a propensity score-matched cohort study. Br J Anaesth. 2022;129:84-91. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 38] [Article Influence: 12.7] [Reference Citation Analysis (0)] |
| 27. | Poly TN, Islam MM, Walther BA, Lin MC, Li YJ. Proton Pump Inhibitors Use and the Risk of Pancreatic Cancer: Evidence from Eleven Epidemiological Studies, Comprising 1.5 Million Individuals. Cancers (Basel). 2022;14:5357. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 8] [Cited by in RCA: 10] [Article Influence: 3.3] [Reference Citation Analysis (0)] |
| 28. | Nagata N, Nishijima S, Kojima Y, Hisada Y, Imbe K, Miyoshi-Akiyama T, Suda W, Kimura M, Aoki R, Sekine K, Ohsugi M, Miki K, Osawa T, Ueki K, Oka S, Mizokami M, Kartal E, Schmidt TSB, Molina-Montes E, Estudillo L, Malats N, Trebicka J, Kersting S, Langheinrich M, Bork P, Uemura N, Itoi T, Kawai T. Metagenomic Identification of Microbial Signatures Predicting Pancreatic Cancer From a Multinational Study. Gastroenterology. 2022;163:222-238. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 46] [Cited by in RCA: 123] [Article Influence: 41.0] [Reference Citation Analysis (0)] |
| 29. | Zamani M, Alizadeh-Tabari S, Murad MH, Ananthakrishnan AN, Malekzadeh R, Talley NJ. Meta-analysis: Risk of pancreatic cancer in patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2024;59:918-927. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 11] [Article Influence: 11.0] [Reference Citation Analysis (0)] |
| 30. | Stoop TF, Javed AA, Oba A, Koerkamp BG, Seufferlein T, Wilmink JW, Besselink MG. Pancreatic cancer. Lancet. 2025;405:1182-1202. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 40] [Article Influence: 40.0] [Reference Citation Analysis (0)] |
| 31. | Heller M, Mann DA, Katona BW. Current Approaches of Pancreatic Cancer Surveillance in High-Risk Individuals. J Gastrointest Cancer. 2025;56:61. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 3] [Reference Citation Analysis (0)] |
| 32. | Del Nero L, Dabizzi E, Ceglie A, Ziola S, Zerbi A, Baron TH, Conio M. Familial pancreatic cancer. Clin Res Hepatol Gastroenterol. 2023;47:102079. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 6] [Reference Citation Analysis (0)] |
| 33. | Bogdanski AM, van Hooft JE, Boekestijn B, Bonsing BA, Wasser MNJM, Klatte DCF, van Leerdam ME. Aspects and outcomes of surveillance for individuals at high-risk of pancreatic cancer. Fam Cancer. 2024;23:323-339. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2] [Cited by in RCA: 6] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
| 34. | Correction: Management of patients with increased risk for familial pancreatic cancer: updated recommendations for the international cancer of the pancreas screening (CAPS) Consortium. Gut. 2020;69:e3. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 11] [Cited by in RCA: 9] [Article Influence: 1.8] [Reference Citation Analysis (0)] |
| 35. | Kogekar N, Diaz KE, Weinberg AD, Lucas AL. Surveillance of high-risk individuals for pancreatic cancer with EUS and MRI: A meta-analysis. Pancreatology. 2020;20:1739-1746. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 14] [Cited by in RCA: 24] [Article Influence: 4.8] [Reference Citation Analysis (0)] |
| 36. | Turner KM, Patel SH. Pancreatic Cancer Screening among High-risk Individuals. Surg Clin North Am. 2024;104:951-964. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 37. | Elbanna KY, Jang HJ, Kim TK. Imaging for Screening/Surveillance of Pancreatic Cancer: A Glimpse of Hope. Korean J Radiol. 2023;24:271-273. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 38. | Aslanian HR, Lee JH, Canto MI. AGA Clinical Practice Update on Pancreas Cancer Screening in High-Risk Individuals: Expert Review. Gastroenterology. 2020;159:358-362. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 92] [Cited by in RCA: 200] [Article Influence: 40.0] [Reference Citation Analysis (0)] |
| 39. | Sawhney MS, Calderwood AH, Thosani NC, Rebbeck TR, Wani S, Canto MI, Fishman DS, Golan T, Hidalgo M, Kwon RS, Riegert-Johnson DL, Sahani DV, Stoffel EM, Vollmer CM Jr, Qumseya BJ; Prepared by: ASGE STANDARDS OF PRACTICE COMMITTEE. ASGE guideline on screening for pancreatic cancer in individuals with genetic susceptibility: summary and recommendations. Gastrointest Endosc. 2022;95:817-826. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 74] [Cited by in RCA: 81] [Article Influence: 27.0] [Reference Citation Analysis (0)] |
| 40. | Zhao B, Zhao B, Chen F. Diagnostic value of serum carbohydrate antigen 19-9 in pancreatic cancer: a systematic review and meta-analysis. Eur J Gastroenterol Hepatol. 2022;34:891-904. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 5] [Cited by in RCA: 16] [Article Influence: 5.3] [Reference Citation Analysis (0)] |
| 41. | Hu W, Zhao X, Luo N, Xiao M, Feng F, An Y, Chen J, Rong L, Yang Y, Peng J. Circulating cell-free DNA methylation analysis of pancreatic cancer patients for early noninvasive diagnosis. Front Oncol. 2025;15:1552426. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
| 42. | Ben-Ami R, Wang QL, Zhang J, Supplee JG, Fahrmann JF, Lehmann-Werman R, Brais LK, Nowak J, Yuan C, Loftus M, Babic A, Irajizad E, Davidi T, Zick A, Hubert A, Neiman D, Piyanzin S, Gal-Rosenberg O, Horn A, Shemer R, Glaser B, Boos N, Jajoo K, Lee L, Clancy TE, Rubinson DA, Ng K, Chabot JA, Kastrinos F, Kluger M, Aguirre AJ, Jänne PA, Bardeesy N, Stanger B, O'Hara MH, Till J, Maitra A, Carpenter EL, Bullock AJ, Genkinger J, Hanash SM, Paweletz CP, Dor Y, Wolpin BM. Protein biomarkers and alternatively methylated cell-free DNA detect early stage pancreatic cancer. Gut. 2024;73:639-648. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 12] [Cited by in RCA: 19] [Article Influence: 19.0] [Reference Citation Analysis (0)] |
| 43. | Madadjim R, An T, Cui J. MicroRNAs in Pancreatic Cancer: Advances in Biomarker Discovery and Therapeutic Implications. Int J Mol Sci. 2024;25:3914. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 26] [Reference Citation Analysis (0)] |
| 44. | Shams R, Saberi S, Zali M, Sadeghi A, Ghafouri-Fard S, Aghdaei HA. Identification of potential microRNA panels for pancreatic cancer diagnosis using microarray datasets and bioinformatics methods. Sci Rep. 2020;10:7559. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 40] [Cited by in RCA: 47] [Article Influence: 9.4] [Reference Citation Analysis (0)] |
| 45. | Huang J, Gao G, Ge Y, Liu J, Cui H, Zheng R, Wang J, Wang S, Go VL, Hu S, Liu Y, Yang M, Sun Y, Shang D, Tian Y, Zhang Z, Xiang Z, Wang H, Guo J, Xiao GG. Development of a Serum-Based MicroRNA Signature for Early Detection of Pancreatic Cancer: A Multicenter Cohort Study. Dig Dis Sci. 2024;69:1263-1273. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 8] [Cited by in RCA: 9] [Article Influence: 9.0] [Reference Citation Analysis (0)] |
| 46. | Yang Z, Huang J, Wu X, Zhou Y, Tang Y, Zhu Y, Li B, Chen X, Yao W. Contribution of a Circulating 2'-O-methylated MicroRNA Panel to the Diagnosis of Pancreatic Ductal Adenocarcinoma. J Cancer. 2024;15:1583-1592. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 5] [Reference Citation Analysis (0)] |
| 47. | Munnings R, Gibbs P, Lee B. Evolution of Liquid Biopsies for Detecting Pancreatic Cancer. Cancers (Basel). 2024;16:3335. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 3] [Reference Citation Analysis (0)] |
| 48. | Wei Q, Wei L, Zhang J, Li Z, Feng H, Ren L. EphA2-enriched exosomes promote cell migration and are a potential diagnostic serum marker in pancreatic cancer. Mol Med Rep. 2020;22:2941-2947. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 4] [Cited by in RCA: 17] [Article Influence: 3.4] [Reference Citation Analysis (0)] |
| 49. | Wei Q, Zhang J, Li Z, Wei L, Ren L. Serum Exo-EphA2 as a Potential Diagnostic Biomarker for Pancreatic Cancer. Pancreas. 2020;49:1213-1219. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 9] [Cited by in RCA: 20] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
| 50. | Xu X, Long C, Li M, Shen C, Ye Q, Li Y, Li H, Cao X, Ma J. Systematic review and meta-analysis: diagnostic accuracy of exosomes in pancreatic cancer. World J Surg Oncol. 2025;23:51. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 2] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
| 51. | Yachida S, Yoshinaga S, Shiba S, Urabe M, Tanaka H, Takeda Y, Shimizu A, Sakamoto Y, Hijioka S, Haba S, Ashida R, Kushiyama Y, Asano K, Kobayashi M, Murawaki Y, Onishi K, Yamashita T, Kimura H, Totoki Y, Kamada H, Isomoto H, Hattori S, Morizane C, Ohkawa K, Kitano M, Hara K, Ikezawa K, Hanada K, Matsumoto K. KRAS Mutations in Duodenal Lavage Fluid after Secretin Stimulation for Detection of Pancreatic Cancer. Ann Surg. 2025. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
| 52. | Sakaue T, Koga H, Iwamoto H, Nakamura T, Masuda A, Tanaka T, Suzuki H, Suga H, Hirai S, Hisaka T, Naito Y, Ohta K, Nakamura KI, Selvendiran K, Okabe Y, Torimura T, Kawaguchi T. Pancreatic Juice-Derived microRNA-4516 and microRNA-4674 as Novel Biomarkers for Pancreatic Ductal Adenocarcinoma. Gastro Hep Adv. 2024;3:761-772. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 3] [Reference Citation Analysis (0)] |
| 53. | Engels MML, Berger CK, Mahoney DW, Hoogenboom SA, Sarwal D, Klatte DCF, De La Fuente J, Gandhi S, Taylor WR, Foote PH, Doering KA, Delgado AM, Burger KN, Abu Dayyeh BK, Bofill-Garcia A, Brahmbhatt B, Chandrasekhara V, Gleeson FC, Gomez V, Kumbhari V, Law RJ, Lukens FJ, Raimondo M, Rajan E, Storm AC, Vargas Valls EJ, van Hooft JE, Wallace MB, Kisiel JB, Majumder S. Multimodal Pancreatic Cancer Detection Using Methylated DNA Biomarkers in Pancreatic Juice and Plasma CA 19-9: A Prospective Multicenter Study. Clin Gastroenterol Hepatol. 2025;23:766-775. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 3] [Reference Citation Analysis (0)] |
| 54. | Conroy T, Pfeiffer P, Vilgrain V, Lamarca A, Seufferlein T, O'Reilly EM, Hackert T, Golan T, Prager G, Haustermans K, Vogel A, Ducreux M; ESMO Guidelines Committee. Pancreatic cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 2023;34:987-1002. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 22] [Cited by in RCA: 254] [Article Influence: 127.0] [Reference Citation Analysis (0)] |
| 55. | van Roessel S, Soer EC, Daamen LA, van Dalen D, Fariña Sarasqueta A, Stommel MWJ, Molenaar IQ, van Santvoort HC, van de Vlasakker VCJ, de Hingh IHJT, Groen JV, Mieog JSD, van Dam JL, van Eijck CHJ, van Tienhoven G, Klümpen HJ, Wilmink JW, Busch OR, Brosens LAA, Groot Koerkamp B, Verheij J, Besselink MG; Dutch Pancreatic Cancer Group. Preoperative misdiagnosis of pancreatic and periampullary cancer in patients undergoing pancreatoduodenectomy: A multicentre retrospective cohort study. Eur J Surg Oncol. 2021;47:2525-2532. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 8] [Cited by in RCA: 31] [Article Influence: 7.8] [Reference Citation Analysis (0)] |
| 56. | Zins M, Matos C, Cassinotto C. Pancreatic Adenocarcinoma Staging in the Era of Preoperative Chemotherapy and Radiation Therapy. Radiology. 2018;287:374-390. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 75] [Cited by in RCA: 119] [Article Influence: 17.0] [Reference Citation Analysis (0)] |
| 57. | Ahmed TM, Chu LC, Javed AA, Yasrab M, Blanco A, Hruban RH, Fishman EK, Kawamoto S. Hidden in plain sight: commonly missed early signs of pancreatic cancer on CT. Abdom Radiol (NY). 2024;49:3599-3614. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 6] [Reference Citation Analysis (0)] |
| 58. | Tempero MA, Malafa MP, Al-Hawary M, Behrman SW, Benson AB, Cardin DB, Chiorean EG, Chung V, Czito B, Del Chiaro M, Dillhoff M, Donahue TR, Dotan E, Ferrone CR, Fountzilas C, Hardacre J, Hawkins WG, Klute K, Ko AH, Kunstman JW, LoConte N, Lowy AM, Moravek C, Nakakura EK, Narang AK, Obando J, Polanco PM, Reddy S, Reyngold M, Scaife C, Shen J, Vollmer C, Wolff RA, Wolpin BM, Lynn B, George GV. Pancreatic Adenocarcinoma, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2021;19:439-457. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 160] [Cited by in RCA: 745] [Article Influence: 186.3] [Reference Citation Analysis (0)] |
| 59. | Raza SS, Khan H, Hajibandeh S, Hajibandeh S, Bartlett D, Chatzizacharias N, Roberts K, Marudanayagam R, Sutcliffe RP. Can preoperative Carbohydrate Antigen 19-9 predict metastatic pancreatic cancer? Results of a systematic review and meta-analysis. HPB (Oxford). 2024;26:630-638. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 8] [Article Influence: 8.0] [Reference Citation Analysis (0)] |
| 60. | Allen PJ, Kuk D, Castillo CF, Basturk O, Wolfgang CL, Cameron JL, Lillemoe KD, Ferrone CR, Morales-Oyarvide V, He J, Weiss MJ, Hruban RH, Gönen M, Klimstra DS, Mino-Kenudson M. Multi-institutional Validation Study of the American Joint Commission on Cancer (8th Edition) Changes for T and N Staging in Patients With Pancreatic Adenocarcinoma. Ann Surg. 2017;265:185-191. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 272] [Cited by in RCA: 364] [Article Influence: 45.5] [Reference Citation Analysis (0)] |
| 61. | Paniccia A, Zureikat A. Editorial on: Moving Beyond Anatomic Criteria for Resectability: Validation of the Anatomical and Biological Definitions of Borderline Resectable Pancreatic Cancer According to the 2017 International Consensus for Survival and Recurrence in Patients with Pancreatic Ductal Adenocarcinoma Undergoing Upfront Surgery. Ann Surg Oncol. 2023;30:3184-3185. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 3] [Article Influence: 1.5] [Reference Citation Analysis (0)] |
| 62. | Isaji S, Mizuno S, Windsor JA, Bassi C, Fernández-Del Castillo C, Hackert T, Hayasaki A, Katz MHG, Kim SW, Kishiwada M, Kitagawa H, Michalski CW, Wolfgang CL. International consensus on definition and criteria of borderline resectable pancreatic ductal adenocarcinoma 2017. Pancreatology. 2018;18:2-11. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 536] [Cited by in RCA: 530] [Article Influence: 75.7] [Reference Citation Analysis (0)] |
| 63. | Nießen A, Hackert T. State-of-the-art surgery for pancreatic cancer. Langenbecks Arch Surg. 2022;407:443-450. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2] [Cited by in RCA: 18] [Article Influence: 4.5] [Reference Citation Analysis (0)] |
| 64. | Shah OJ, Singh M. Developments in pancreatic cancer surgery. Updates Surg. 2024;76:17-22. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 6] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
| 65. | Palanivelu C, Senthilnathan P, Sabnis SC, Babu NS, Srivatsan Gurumurthy S, Anand Vijai N, Nalankilli VP, Praveen Raj P, Parthasarathy R, Rajapandian S. Randomized clinical trial of laparoscopic versus open pancreatoduodenectomy for periampullary tumours. Br J Surg. 2017;104:1443-1450. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 196] [Cited by in RCA: 302] [Article Influence: 37.8] [Reference Citation Analysis (0)] |
| 66. | Poves I, Burdío F, Morató O, Iglesias M, Radosevic A, Ilzarbe L, Visa L, Grande L. Comparison of Perioperative Outcomes Between Laparoscopic and Open Approach for Pancreatoduodenectomy: The PADULAP Randomized Controlled Trial. Ann Surg. 2018;268:731-739. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 166] [Cited by in RCA: 278] [Article Influence: 46.3] [Reference Citation Analysis (0)] |
| 67. | van Hilst J, de Rooij T, Bosscha K, Brinkman DJ, van Dieren S, Dijkgraaf MG, Gerhards MF, de Hingh IH, Karsten TM, Lips DJ, Luyer MD, Busch OR, Festen S, Besselink MG; Dutch Pancreatic Cancer Group. Laparoscopic versus open pancreatoduodenectomy for pancreatic or periampullary tumours (LEOPARD-2): a multicentre, patient-blinded, randomised controlled phase 2/3 trial. Lancet Gastroenterol Hepatol. 2019;4:199-207. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 429] [Cited by in RCA: 426] [Article Influence: 71.0] [Reference Citation Analysis (0)] |
| 68. | Nickel F, Haney CM, Kowalewski KF, Probst P, Limen EF, Kalkum E, Diener MK, Strobel O, Müller-Stich BP, Hackert T. Laparoscopic Versus Open Pancreaticoduodenectomy: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Ann Surg. 2020;271:54-66. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 106] [Cited by in RCA: 202] [Article Influence: 40.4] [Reference Citation Analysis (0)] |
| 69. | Choi SH, Kuchta K, Rojas AE, Paterakos P, Talamonti MS, Hogg ME. Does minimally invasive surgery have a different impact on recurrence and overall survival in patients with pancreatic head versus body/tail cancer? J Surg Oncol. 2023;128:23-32. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 70. | Giani A, Mazzola M, Paterno M, Zironda A, Calcagno P, Zuppi E, De Martini P, Ferrari G. Oncological Outcomes of Open Versus Minimally Invasive Surgery for Ductal Adenocarcinomas of Pancreatic Head: A Propensity Score Matching Analysis. Curr Oncol. 2024;31:6096-6109. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 2] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
| 71. | Nassour I, Winters SB, Hoehn R, Tohme S, Adam MA, Bartlett DL, Lee KK, Paniccia A, Zureikat AH. Long-term oncologic outcomes of robotic and open pancreatectomy in a national cohort of pancreatic adenocarcinoma. J Surg Oncol. 2020;122:234-242. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 30] [Cited by in RCA: 55] [Article Influence: 11.0] [Reference Citation Analysis (0)] |
| 72. | Nassour I, Tohme S, Hoehn R, Adam MA, Zureikat AH, Alessandro P. Safety and oncologic efficacy of robotic compared to open pancreaticoduodenectomy after neoadjuvant chemotherapy for pancreatic cancer. Surg Endosc. 2021;35:2248-2254. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 12] [Cited by in RCA: 23] [Article Influence: 4.6] [Reference Citation Analysis (0)] |
| 73. | Girgis MD, Zenati MS, King JC, Hamad A, Zureikat AH, Zeh HJ, Hogg ME. Oncologic Outcomes After Robotic Pancreatic Resections Are Not Inferior to Open Surgery. Ann Surg. 2021;274:e262-e268. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 28] [Cited by in RCA: 56] [Article Influence: 9.3] [Reference Citation Analysis (0)] |
| 74. | Klotz R, Mihaljevic AL, Kulu Y, Sander A, Klose C, Behnisch R, Joos MC, Kalkum E, Nickel F, Knebel P, Pianka F, Diener MK, Büchler MW, Hackert T. Robotic versus open partial pancreatoduodenectomy (EUROPA): a randomised controlled stage 2b trial. Lancet Reg Health Eur. 2024;39:100864. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 4] [Cited by in RCA: 49] [Article Influence: 49.0] [Reference Citation Analysis (0)] |
| 75. | Mosalem OM, Abdelhakeem A, Abdel-Razeq NH, Babiker H. Pancreatic ductal adenocarcinoma (PDAC): clinical progress in the last five years. Expert Opin Investig Drugs. 2025;34:149-160. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 7] [Reference Citation Analysis (0)] |
| 76. | Dai M, Chen L, Xu Q, Cui M, Li P, Liu W, Lin C, Chen W, Chen H, Yuan S. Robotic Versus Laparoscopic Pancreaticoduodenectomy for Pancreatic Cancer: Evaluation and Analysis of Surgical Efficacy. Ann Surg Oncol. 2024;31:7043-7051. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 9] [Article Influence: 9.0] [Reference Citation Analysis (0)] |
| 77. | de Rooij T, van Hilst J, van Santvoort H, Boerma D, van den Boezem P, Daams F, van Dam R, Dejong C, van Duyn E, Dijkgraaf M, van Eijck C, Festen S, Gerhards M, Groot Koerkamp B, de Hingh I, Kazemier G, Klaase J, de Kleine R, van Laarhoven C, Luyer M, Patijn G, Steenvoorde P, Suker M, Abu Hilal M, Busch O, Besselink M; Dutch Pancreatic Cancer Group. Minimally Invasive Versus Open Distal Pancreatectomy (LEOPARD): A Multicenter Patient-blinded Randomized Controlled Trial. Ann Surg. 2019;269:2-9. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 460] [Cited by in RCA: 420] [Article Influence: 70.0] [Reference Citation Analysis (0)] |
| 78. | van Hilst J, de Rooij T, Klompmaker S, Rawashdeh M, Aleotti F, Al-Sarireh B, Alseidi A, Ateeb Z, Balzano G, Berrevoet F, Björnsson B, Boggi U, Busch OR, Butturini G, Casadei R, Del Chiaro M, Chikhladze S, Cipriani F, van Dam R, Damoli I, van Dieren S, Dokmak S, Edwin B, van Eijck C, Fabre JM, Falconi M, Farges O, Fernández-Cruz L, Forgione A, Frigerio I, Fuks D, Gavazzi F, Gayet B, Giardino A, Groot Koerkamp B, Hackert T, Hassenpflug M, Kabir I, Keck T, Khatkov I, Kusar M, Lombardo C, Marchegiani G, Marshall R, Menon KV, Montorsi M, Orville M, de Pastena M, Pietrabissa A, Poves I, Primrose J, Pugliese R, Ricci C, Roberts K, Røsok B, Sahakyan MA, Sánchez-Cabús S, Sandström P, Scovel L, Solaini L, Soonawalla Z, Souche FR, Sutcliffe RP, Tiberio GA, Tomazic A, Troisi R, Wellner U, White S, Wittel UA, Zerbi A, Bassi C, Besselink MG, Abu Hilal M; European Consortium on Minimally Invasive Pancreatic Surgery (E-MIPS). Minimally Invasive versus Open Distal Pancreatectomy for Ductal Adenocarcinoma (DIPLOMA): A Pan-European Propensity Score Matched Study. Ann Surg. 2019;269:10-17. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 227] [Cited by in RCA: 205] [Article Influence: 34.2] [Reference Citation Analysis (0)] |
| 79. | Korrel M, Jones LR, van Hilst J, Balzano G, Björnsson B, Boggi U, Bratlie SO, Busch OR, Butturini G, Capretti G, Casadei R, Edwin B, Emmen AMLH, Esposito A, Falconi M, Groot Koerkamp B, Keck T, de Kleine RHJ, Kleive DB, Kokkola A, Lips DJ, Lof S, Luyer MDP, Manzoni A, Marudanayagam R, de Pastena M, Pecorelli N, Primrose JN, Ricci C, Salvia R, Sandström P, Vissers FLIM, Wellner UF, Zerbi A, Dijkgraaf MGW, Besselink MG, Abu Hilal M; European Consortium on Minimally Invasive Pancreatic Surgery (E-MIPS). Minimally invasive versus open distal pancreatectomy for resectable pancreatic cancer (DIPLOMA): an international randomised non-inferiority trial. Lancet Reg Health Eur. 2023;31:100673. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 49] [Cited by in RCA: 67] [Article Influence: 33.5] [Reference Citation Analysis (0)] |
| 80. | Hays SB, Rojas AE, Hogg ME. Robotic pancreas surgery for pancreatic cancer. Int J Surg. 2024;110:6100-6110. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 5] [Cited by in RCA: 5] [Article Influence: 5.0] [Reference Citation Analysis (0)] |
| 81. | van Bodegraven EA, van Ramshorst TME, Bratlie SO, Kokkola A, Sparrelid E, Björnsson B, Kleive D, Burgdorf SK, Dokmak S, Groot Koerkamp B, Cabús SS, Molenaar IQ, Boggi U, Busch OR, Petrič M, Roeyen G, Hackert T, Lips DJ, D'Hondt M, Coolsen MME, Ferrari G, Tingstedt B, Serrablo A, Gaujoux S, Ramera M, Khatkov I, Ausania F, Souche R, Festen S, Berrevoet F, Keck T, Sutcliffe RP, Pando E, de Wilde RF, Aussilhou B, Krohn PS, Edwin B, Sandström P, Gilg S, Seppänen H, Vilhav C, Abu Hilal M, Besselink MG; European Consortium on Minimally Invasive Pancreatic Surgery (E-MIPS). Minimally invasive robot-assisted and laparoscopic distal pancreatectomy in a pan-European registry a retrospective cohort study. Int J Surg. 2024;110:3554-3561. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 6] [Cited by in RCA: 6] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
| 82. | Chen JW, van Ramshorst TME, Lof S, Al-Sarireh B, Bjornsson B, Boggi U, Burdio F, Butturini G, Casadei R, Coratti A, D'Hondt M, Dokmak S, Edwin B, Esposito A, Fabre JM, Ferrari G, Ftériche FS, Fusai GK, Groot Koerkamp B, Hackert T, Jah A, Jang JY, Kauffmann EF, Keck T, Manzoni A, Marino MV, Molenaar Q, Pando E, Pessaux P, Pietrabissa A, Soonawalla Z, Sutcliffe RP, Timmermann L, White S, Yip VS, Zerbi A, Abu Hilal M, Besselink MG; European Consortium on Minimally Invasive Pancreatic Surgery (E-MIPS). Robot-Assisted Versus Laparoscopic Distal Pancreatectomy in Patients with Resectable Pancreatic Cancer: An International, Retrospective, Cohort Study. Ann Surg Oncol. 2023;30:3023-3032. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 23] [Reference Citation Analysis (0)] |
| 83. | Concors SJ, Katz MHG, Ikoma N. Minimally Invasive Pancreatectomy: Robotic and Laparoscopic Developments. Surg Oncol Clin N Am. 2023;32:327-342. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 4] [Reference Citation Analysis (0)] |
| 84. | Scholten L, Klompmaker S, Van Hilst J, Annecchiarico MM, Balzano G, Casadei R, Fabre JM, Falconi M, Ferrari G, Kerem M, Khatkov IE, Lombardo C, Manzoni A, Mazzola M, Napoli N, Rosso EE, Tyutyunnik P, Wellner UF, Fuks D, Burdio F, Keck T, Hilal MA, Besselink MG, Boggi U; European consortium on Minimally Invasive Pancreatic Surgery and the Scientific and Research Committee of the European-African Hepato-Pancreato-Biliary Association. Outcomes After Minimally Invasive Versus Open Total Pancreatectomy: A Pan-European Propensity Score Matched Study. Ann Surg. 2023;277:313-320. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 8] [Reference Citation Analysis (0)] |
| 85. | Bengtsson A, Andersson R, Ansari D. The actual 5-year survivors of pancreatic ductal adenocarcinoma based on real-world data. Sci Rep. 2020;10:16425. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 260] [Cited by in RCA: 282] [Article Influence: 56.4] [Reference Citation Analysis (0)] |
| 86. | Daamen LA, Dorland G, Brada LJH, Groot VP, van Oosten AF, Besselink MG, Bosscha K, Bonsing BA, Busch OR, Cirkel GA, van Dam RM, Festen S, Groot Koerkamp B, Haj Mohammad N, van der Harst E, de Hingh IHJT, Intven MPW, Kazemier G, Los M, de Meijer VE, Nieuwenhuijs VB, Roos D, Schreinemakers JMJ, Stommel MWJ, Verdonk RC, Verkooijen HM, Molenaar IQ, van Santvoort HC; Dutch Pancreatic Cancer Group. Preoperative predictors for early and very early disease recurrence in patients undergoing resection of pancreatic ductal adenocarcinoma. HPB (Oxford). 2022;24:535-546. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 8] [Cited by in RCA: 27] [Article Influence: 9.0] [Reference Citation Analysis (0)] |
| 87. | van Goor IWJM, Schouten TJ, Verburg DN, Besselink MG, Bonsing BA, Bosscha K, Brosens LAA, Busch OR, Cirkel GA, van Dam RM, Festen S, Koerkamp BG, van der Harst E, de Hingh IHJT, Intven MPW, Kazemier G, Los M, Meijer GJ, de Meijer VE, Nieuwenhuijs VB, Roos D, Schreinemakers JMJ, Stommel MWJ, Verdonk RC, van Santvoort HC, Daamen LA, Molenaar IQ; Dutch Pancreatic Cancer Group. Predicting Long-term Disease-free Survival After Resection of Pancreatic Ductal Adenocarcinoma: A Nationwide Cohort Study. Ann Surg. 2024;279:132-137. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 7] [Cited by in RCA: 15] [Article Influence: 15.0] [Reference Citation Analysis (0)] |
| 88. | Koti S, Demyan L, Deutsch G, Weiss M. Surgery for Oligometastatic Pancreatic Cancer: Defining Biologic Resectability. Ann Surg Oncol. 2024;31:4031-4041. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 9] [Reference Citation Analysis (0)] |
| 89. | Lievens Y, Guckenberger M, Gomez D, Hoyer M, Iyengar P, Kindts I, Méndez Romero A, Nevens D, Palma D, Park C, Ricardi U, Scorsetti M, Yu J, Woodward WA. Defining oligometastatic disease from a radiation oncology perspective: An ESTRO-ASTRO consensus document. Radiother Oncol. 2020;148:157-166. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 162] [Cited by in RCA: 451] [Article Influence: 90.2] [Reference Citation Analysis (0)] |
| 90. | Leonhardt CS, Stamm T, Hank T, Prager G, Strobel O. Defining oligometastatic pancreatic cancer: a systematic review and critical synthesis of consensus. ESMO Open. 2023;8:102067. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 20] [Cited by in RCA: 14] [Article Influence: 7.0] [Reference Citation Analysis (0)] |
| 91. | Horn SR, Stoltzfus KC, Lehrer EJ, Dawson LA, Tchelebi L, Gusani NJ, Sharma NK, Chen H, Trifiletti DM, Zaorsky NG. Epidemiology of liver metastases. Cancer Epidemiol. 2020;67:101760. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 45] [Cited by in RCA: 184] [Article Influence: 36.8] [Reference Citation Analysis (0)] |
| 92. | Frigerio I, Malleo G, de Pastena M, Deiro G, Surci N, Scopelliti F, Esposito A, Regi P, Giardino A, Allegrini V, Bassi C, Girelli R, Salvia R, Butturini G. Prognostic Factors After Pancreatectomy for Pancreatic Cancer Initially Metastatic to the Liver. Ann Surg Oncol. 2022;29:8503-8510. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 20] [Reference Citation Analysis (0)] |
| 93. | Bachellier P, Addeo P, Averous G, Dufour P. Resection of pancreatic adenocarcinomas with synchronous liver metastases: A retrospective study of prognostic factors for survival. Surgery. 2022;172:1245-1250. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 9] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
| 94. | Homma Y, Endo I, Matsuyama R, Sho M, Mizuno S, Seyama Y, Hirano S, Aono T, Kitami C, Morita Y, Takeda Y, Yoshida K, Tani M, Kaiho T, Yamamoto Y, Aoki H, Ogawa M, Niguma T, Mataki Y, Kawasaki H, Baba H, Yokomizo H, Rikiyama T, Yamaue H, Yamamoto M. Outcomes of lung metastasis from pancreatic cancer: A nationwide multicenter analysis. J Hepatobiliary Pancreat Sci. 2022;29:552-561. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 32] [Cited by in RCA: 32] [Article Influence: 10.7] [Reference Citation Analysis (0)] |
| 95. | Shimizu T, Taniguchi K, Asakuma M, Komeda K, Inoue Y, Lee SW, Hirokawa F, Uchiyama K. Initial pulmonary metastasis after pancreatectomy for pancreatic ductal adenocarcinoma. Surg Today. 2020;50:413-418. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 9] [Article Influence: 1.5] [Reference Citation Analysis (0)] |
| 96. | Yasukawa M, Kawaguchi T, Kawai N, Tojo T, Taniguchi S. Surgical Treatment for Pulmonary Metastasis of Pancreatic Ductal Adenocarcinoma: Study of 12 Cases. Anticancer Res. 2017;37:5573-5576. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3] [Cited by in RCA: 8] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
| 97. | Kaiho T, Suzuki H, Yamamoto T, Morimoto J, Sakairi Y, Wada H, Nakajima T, Yoshino I. Surgical outcomes of pulmonary metastasis from hepatopancreatobiliary carcinomas: a comparison with pulmonary metastasis from colorectal carcinomas. Surg Today. 2019;49:762-768. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Cited by in RCA: 6] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
| 98. | Ilmer M, Schiergens TS, Renz BW, Schneider C, Sargut M, Waligora R, Weniger M, Hartwig W, Ceyhan GO, Friess H, Werner J, D'Haese JG. Oligometastatic pulmonary metastasis in pancreatic cancer patients: Safety and outcome of resection. Surg Oncol. 2019;31:16-21. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 21] [Cited by in RCA: 44] [Article Influence: 7.3] [Reference Citation Analysis (0)] |
| 99. | Yun WG, Kwon W, Han Y, Sohn HJ, Kim HS, Lee M, Kim H, Thomas AS, Kluger MD, Jang JY. Can Surgical Resection of Metastatic Lesions Be Beneficial to Pancreatic Ductal Adenocarcinoma Patients with Isolated Lung Metastasis? Cancers (Basel). 2022;14:2067. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2] [Cited by in RCA: 11] [Article Influence: 3.7] [Reference Citation Analysis (0)] |
| 100. | Yan G, Zhang K, Yan L, Zhang Y. Efficacy and safety of cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy in patients with pancreatic cancer peritoneal metastasis. World J Surg Oncol. 2024;22:212. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 4] [Reference Citation Analysis (0)] |
| 101. | Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN, Harris M, Reni M, Dowden S, Laheru D, Bahary N, Ramanathan RK, Tabernero J, Hidalgo M, Goldstein D, Van Cutsem E, Wei X, Iglesias J, Renschler MF. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691-1703. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4035] [Cited by in RCA: 4997] [Article Influence: 416.4] [Reference Citation Analysis (7)] |
| 102. | Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardière C, Bennouna J, Bachet JB, Khemissa-Akouz F, Péré-Vergé D, Delbaldo C, Assenat E, Chauffert B, Michel P, Montoto-Grillot C, Ducreux M; Groupe Tumeurs Digestives of Unicancer; PRODIGE Intergroup. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817-1825. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4838] [Cited by in RCA: 5752] [Article Influence: 410.9] [Reference Citation Analysis (4)] |
| 103. | Di Costanzo F, Di Costanzo F, Antonuzzo L, Mazza E, Giommoni E. Optimizing First-Line Chemotherapy in Metastatic Pancreatic Cancer: Efficacy of FOLFIRINOX versus Nab-Paclitaxel Plus Gemcitabine. Cancers (Basel). 2023;15:416. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 28] [Reference Citation Analysis (0)] |
| 104. | Frassini S, Calabretto F, Granieri S, Fugazzola P, Viganò J, Fazzini N, Ansaloni L, Cobianchi L. Intraperitoneal chemotherapy in the management of pancreatic adenocarcinoma: A systematic review and meta-analysis. Eur J Surg Oncol. 2022;48:1911-1921. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3] [Cited by in RCA: 15] [Article Influence: 5.0] [Reference Citation Analysis (0)] |
| 105. | Safari D, Fakhrolmobasheri M, Soleymanjahi S. Efficacy and safety of intraperitoneal chemotherapy for pancreatic cancer. BMC Surg. 2024;24:285. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
| 106. | Yamamoto T, Satoi S, Yamaki S, Hashimoto D, Ishida M, Ikeura T, Hirooka S, Matsui Y, Boku S, Nakayama S, Nakamaru K, Shibata N, Katsushima U, Sekimoto M. Intraperitoneal Paclitaxel Treatment for Patients with Pancreatic Ductal Adenocarcinoma with Peritoneal Dissemination Provides a Survival Benefit. Cancers (Basel). 2022;14:1354. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 3] [Cited by in RCA: 12] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
| 107. | Gudmundsdottir H, Yonkus JA, Thiels CA, Warner SG, Cleary SP, Kendrick ML, Truty MJ, Grotz TE. Oncologic Outcomes of Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy for Highly Selected Patients with Metastatic Pancreatic Ductal Adenocarcinoma. Ann Surg Oncol. 2023;30:7833-7839. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 8] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
| 108. | Tentes AA, Pallas N, Karamveri C, Kyziridis D, Hristakis C. Cytoreduction and HIPEC for peritoneal carcinomatosis of pancreatic cancer. J BUON. 2018;23:482-487. [PubMed] |
| 109. | Tentes AA, Kyziridis D, Kalakonas A, Iliadis A, Fotiadou A. Pancreatic cancer with synchronous peritoneal and hepatic metastases: A case report. Int J Surg Case Rep. 2024;118:109588. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 110. | Graversen M, Detlefsen S, Ainsworth AP, Fristrup CW, Knudsen AO, Pfeiffer P, Tarpgaard LS, Mortensen MB. Treatment of Peritoneal Metastasis with Pressurized Intraperitoneal Aerosol Chemotherapy: Results from the Prospective PIPAC-OPC2 Study. Ann Surg Oncol. 2023;30:2634-2644. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 33] [Reference Citation Analysis (0)] |
| 111. | Di Giorgio A, Sgarbura O, Rotolo S, Schena CA, Bagalà C, Inzani F, Russo A, Chiantera V, Pacelli F. Pressurized intraperitoneal aerosol chemotherapy with cisplatin and doxorubicin or oxaliplatin for peritoneal metastasis from pancreatic adenocarcinoma and cholangiocarcinoma. Ther Adv Med Oncol. 2020;12:1758835920940887. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 8] [Cited by in RCA: 20] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
| 112. | Ceelen W, Sandra L, de Sande LV, Graversen M, Mortensen MB, Vermeulen A, Gasthuys E, Reynders D, Cosyns S, Hoorens A, Willaert W. Phase I study of intraperitoneal aerosolized nanoparticle albumin based paclitaxel (NAB-PTX) for unresectable peritoneal metastases. EBioMedicine. 2022;82:104151. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 31] [Cited by in RCA: 27] [Article Influence: 9.0] [Reference Citation Analysis (0)] |
| 113. | de Jong LAW, van Erp NP, Bijelic L. Pressurized Intraperitoneal Aerosol Chemotherapy: The Road from Promise to Proof. Clin Cancer Res. 2021;27:1830-1832. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 6] [Article Influence: 1.5] [Reference Citation Analysis (0)] |
| 114. | Hu ZI, O'Reilly EM. Therapeutic developments in pancreatic cancer. Nat Rev Gastroenterol Hepatol. 2024;21:7-24. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 107] [Cited by in RCA: 197] [Article Influence: 197.0] [Reference Citation Analysis (0)] |
| 115. | Conroy T, Castan F, Lopez A, Turpin A, Ben Abdelghani M, Wei AC, Mitry E, Biagi JJ, Evesque L, Artru P, Lecomte T, Assenat E, Bauguion L, Ychou M, Bouché O, Monard L, Lambert A, Hammel P; Canadian Cancer Trials Group and the Unicancer-GI–PRODIGE Group. Five-Year Outcomes of FOLFIRINOX vs Gemcitabine as Adjuvant Therapy for Pancreatic Cancer: A Randomized Clinical Trial. JAMA Oncol. 2022;8:1571-1578. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 41] [Cited by in RCA: 205] [Article Influence: 68.3] [Reference Citation Analysis (0)] |
| 116. | Uesaka K, Boku N, Fukutomi A, Okamura Y, Konishi M, Matsumoto I, Kaneoka Y, Shimizu Y, Nakamori S, Sakamoto H, Morinaga S, Kainuma O, Imai K, Sata N, Hishinuma S, Ojima H, Yamaguchi R, Hirano S, Sudo T, Ohashi Y; JASPAC 01 Study Group. Adjuvant chemotherapy of S-1 versus gemcitabine for resected pancreatic cancer: a phase 3, open-label, randomised, non-inferiority trial (JASPAC 01). Lancet. 2016;388:248-257. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 635] [Cited by in RCA: 824] [Article Influence: 91.6] [Reference Citation Analysis (0)] |
| 117. | Mackay TM, Smits FJ, Roos D, Bonsing BA, Bosscha K, Busch OR, Creemers GJ, van Dam RM, van Eijck CHJ, Gerhards MF, de Groot JWB, Groot Koerkamp B, Haj Mohammad N, van der Harst E, de Hingh IHJT, Homs MYV, Kazemier G, Liem MSL, de Meijer VE, Molenaar IQ, Nieuwenhuijs VB, van Santvoort HC, van der Schelling GP, Stommel MWJ, Ten Tije AJ, de Vos-Geelen J, Wit F, Wilmink JW, van Laarhoven HWM, Besselink MG; Dutch Pancreatic Cancer Group. The risk of not receiving adjuvant chemotherapy after resection of pancreatic ductal adenocarcinoma: a nationwide analysis. HPB (Oxford). 2020;22:233-240. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 36] [Cited by in RCA: 78] [Article Influence: 15.6] [Reference Citation Analysis (0)] |
| 118. | Aliseda D, Martí-Cruchaga P, Zozaya G, Blanco N, Ponz M, Chopitea A, Rodríguez J, Castañón E, Pardo F, Rotellar F. Neoadjuvant therapy versus upfront surgery in resectable pancreatic cancer: reconstructed patient-level meta-analysis of randomized clinical trials. BJS Open. 2024;8:zrae087. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 10] [Reference Citation Analysis (0)] |
| 119. | Crippa S, Malleo G, Mazzaferro V, Langella S, Ricci C, Casciani F, Belfiori G, Galati S, D'Ambra V, Lionetto G, Ferrero A, Casadei R, Ercolani G, Salvia R, Falconi M, Cucchetti A. Futility of Up-Front Resection for Anatomically Resectable Pancreatic Cancer. JAMA Surg. 2024;159:1139-1147. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 24] [Reference Citation Analysis (0)] |
| 120. | Rangelova E, Stoop TF, van Ramshorst TME, Ali M, van Bodegraven EA, Javed AA, Hashimoto D, Steyerberg E, Banerjee A, Jain A, Sauvanet A, Serrablo A, Giani A, Giardino A, Zerbi A, Arshad A, Wijma AG, Coratti A, Zironda A, Socratous A, Rojas A, Halimi A, Ejaz A, Oba A, Patel BY, Björnsson B, Reames BN, Tingstedt B, Goh BKP, Payá-Llorente C, Del Pozo CD, González-Abós C, Medin C, van Eijck CHJ, de Ponthaud C, Takishita C, Schwabl C, Månsson C, Ricci C, Thiels CA, Douchi D, Hughes DL, Kilburn D, Flanking D, Kleive D, Silva DS, Edil BH, Pando E, Moltzer E, Kauffman EF, Warren E, Bozkurt E, Sparrelid E, Thoma E, Verkolf E, Ausania F, Giannone F, Hüttner FJ, Burdio F, Souche FR, Berrevoet F, Daams F, Motoi F, Saliba G, Kazemier G, Roeyen G, Nappo G, Butturini G, Ferrari G, Kito Fusai G, Honda G, Sergeant G, Karteszi H, Takami H, Suto H, Matsumoto I, Mora-Oliver I, Frigerio I, Fabre JM, Chen J, Sham JG, Davide J, Urdzik J, de Martino J, Nielsen K, Okano K, Kamei K, Okada K, Tanaka K, Labori KJ, Goodsell KE, Alberici L, Webber L, Kirkov L, de Franco L, Miyashita M, Maglione M, Gramellini M, Ramera M, Amaral MJ, Ramaekers M, Truty MJ, van Dam MA, Stommel MWJ, Petrikowski M, Imamura M, Hayashi M, D'Hondt M, Brunner M, Hogg ME, Zhang C, Suárez-Muñoz MÁ, Luyer MD, Unno M, Mizuma M, Janot M, Sahakyan MA, Jamieson NB, Busch OR, Bilge O, Belyaev O, Franklin O, Sánchez-Velázquez P, Pessaux P, Holka PS, Ghorbani P, Casadei R, Sartoris R, Schulick RD, Grützmann R, Sutcliffe R, Mata R, Patel RB, Takahashi R, Rodriguez Franco S, Cabús SS, Hirano S, Gaujoux S, Festen S, Kozono S, Maithel SK, Chai SM, Yamaki S, van Laarhoven S, Mieog JSD, Murakami T, Codjia T, Sumiyoshi T, Karsten TM, Nakamura T, Sugawara T, Boggi U, Hartman V, de Meijer VE, Bartholomä W, Kwon W, Koh YX, Cho Y, Takeyama Y, Inoue Y, Nagakawa Y, Kawamoto Y, Ome Y, Soonawalla Z, Uemura K, Wolfgang CL, Jang JY, Padbury R, Satoi S, Messersmith W, Wilmink JW, Abu Hilal M, Besselink MG, Del Chiaro M; European Consortium on Minimally Invasive Pancreatic Surgery (E-MIPS); International Consortium on Advanced Pancreatic Surgery. The impact of neoadjuvant therapy in patients with left-sided resectable pancreatic cancer: an international multicenter study. Ann Oncol. 2025;36:529-542. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 5] [Reference Citation Analysis (0)] |
| 121. | Brown ZJ, Heh V, Labiner HE, Brock GN, Ejaz A, Dillhoff M, Tsung A, Pawlik TM, Cloyd JM. Surgical resection rates after neoadjuvant therapy for localized pancreatic ductal adenocarcinoma: meta-analysis. Br J Surg. 2022;110:34-42. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 58] [Reference Citation Analysis (0)] |
| 122. | Janssen QP, van Dam JL, Bonsing BA, Bos H, Bosscha KP, Coene PPLO, van Eijck CHJ, de Hingh IHJT, Karsten TM, van der Kolk MB, Patijn GA, Liem MSL, van Santvoort HC, Loosveld OJL, de Vos-Geelen J, Zonderhuis BM, Homs MYV, van Tienhoven G, Besselink MG, Wilmink JW, Groot Koerkamp B; Dutch Pancreatic Cancer Group. Total neoadjuvant FOLFIRINOX versus neoadjuvant gemcitabine-based chemoradiotherapy and adjuvant gemcitabine for resectable and borderline resectable pancreatic cancer (PREOPANC-2 trial): study protocol for a nationwide multicenter randomized controlled trial. BMC Cancer. 2021;21:300. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 83] [Cited by in RCA: 130] [Article Influence: 32.5] [Reference Citation Analysis (0)] |
| 123. | Ikeda M, Nomura S, Kobayashi T, Kimura Y, Morinaga S, Toyama H, Sugiura T, Hirano S, Shimizu Y, Tomikawa M, Sadamori H, Katanuma A, Horie Y, Unno M, Sugimachi K, Yamaguchi H, Kojima M, Akimoto T, Uesaka K, Takahashi S. Randomized phase II/III trial of gemcitabine plus nab-paclitaxel versus concurrent chemoradiotherapy with S-1 as neoadjuvant treatment for borderline resectable pancreatic cancer: GABARNANCE study. J Clin Oncol. 2024;42:LBA4014-LBA4014. [DOI] [Full Text] |
| 124. | Lambert A, Bouche O, Ayav A, Bachet J, Schwarz L, Piessen G, Vendrely V, Laurent V, Thibaudeau E, Miglianico L, Rinaldi Y, Hammel P, Conroy T. LBA62 Preoperative modified FOLFIRINOX (mFOLFIRINOX) with or without chemoradiation (CRT) in borderline resectable pancreatic cancer (BRPC): Results from the randomized phase II trial PANDAS/PRODIGE 44. Ann Oncol. 2024;35:S1252. [DOI] [Full Text] |
| 125. | Yun WG, Chae YS, Han Y, Jung HS, Cho YJ, Kang HC, Kwon W, Park JS, Chie EK, Jang JY. Efficacy of Neoadjuvant Radiotherapy After Chemotherapy and the Optimal Interval from Radiotherapy to Surgery for Borderline Resectable and Resectable Pancreatic Cancer. Ann Surg Oncol. 2025;32:2819-2829. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Cited by in RCA: 6] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
| 126. | Sohal DPS, Duong M, Ahmad SA, Gandhi NS, Beg MS, Wang-Gillam A, Wade JL 3rd, Chiorean EG, Guthrie KA, Lowy AM, Philip PA, Hochster HS. Efficacy of Perioperative Chemotherapy for Resectable Pancreatic Adenocarcinoma: A Phase 2 Randomized Clinical Trial. JAMA Oncol. 2021;7:421-427. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 156] [Cited by in RCA: 212] [Article Influence: 53.0] [Reference Citation Analysis (0)] |
| 127. | Yamaguchi J, Yokoyama Y, Fujii T, Yamada S, Takami H, Kawashima H, Ohno E, Ishikawa T, Maeda O, Ogawa H, Kodera Y, Nagino M, Ebata T. Results of a Phase II Study on the Use of Neoadjuvant Chemotherapy (FOLFIRINOX or GEM/nab-PTX) for Borderline-resectable Pancreatic Cancer (NUPAT-01). Ann Surg. 2022;275:1043-1049. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 68] [Cited by in RCA: 65] [Article Influence: 21.7] [Reference Citation Analysis (0)] |
| 128. | Ozaka M, Nakachi K, Kobayashi S, Ohba A, Imaoka H, Terashima T, Ishii H, Mizusawa J, Katayama H, Kataoka T, Okusaka T, Ikeda M, Sasahira N, Miwa H, Mizukoshi E, Okano N, Mizuno N, Yamamoto T, Komatsu Y, Todaka A, Kamata K, Furukawa M, Fujimori N, Katanuma A, Takayama Y, Tsumura H, Fukuda H, Ueno M, Furuse J; Hepatobiliary and Pancreatic Oncology Group of Japan Clinical Oncology Group (JCOG). A randomised phase II study of modified FOLFIRINOX versus gemcitabine plus nab-paclitaxel for locally advanced pancreatic cancer (JCOG1407). Eur J Cancer. 2023;181:135-144. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 51] [Reference Citation Analysis (0)] |
| 129. | Tchelebi LT, Lehrer EJ, Trifiletti DM, Sharma NK, Gusani NJ, Crane CH, Zaorsky NG. Conventionally fractionated radiation therapy versus stereotactic body radiation therapy for locally advanced pancreatic cancer (CRiSP): An international systematic review and meta-analysis. Cancer. 2020;126:2120-2131. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 40] [Cited by in RCA: 92] [Article Influence: 18.4] [Reference Citation Analysis (0)] |
| 130. | Janssen QP, van Dam JL, Kivits IG, Besselink MG, van Eijck CHJ, Homs MYV, Nuyttens JJME, Qi H, van Santvoort HJ, Wei AC, de Wilde RF, Wilmink JW, van Tienhoven G, Groot Koerkamp B. Added Value of Radiotherapy Following Neoadjuvant FOLFIRINOX for Resectable and Borderline Resectable Pancreatic Cancer: A Systematic Review and Meta-Analysis. Ann Surg Oncol. 2021;28:8297-8308. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 6] [Cited by in RCA: 30] [Article Influence: 7.5] [Reference Citation Analysis (0)] |
| 131. | Stoop TF, Oba A, Wu YHA, Beaty LE, Colborn KL, Janssen BV, Al-Musawi MH, Franco SR, Sugawara T, Franklin O, Jain A, Saiura A, Sauvanet A, Coppola A, Javed AA, Groot Koerkamp B, Miller BN, Mack CE, Hashimoto D, Caputo D, Kleive D, Sereni E, Belfiori G, Ichida H, van Dam JL, Dembinski J, Akahoshi K, Roberts KJ, Tanaka K, Labori KJ, Falconi M, House MG, Sugimoto M, Tanabe M, Gotohda N, Krohn PS, Burkhart RA, Thakkar RG, Pande R, Dokmak S, Hirano S, Burgdorf SK, Crippa S, van Roessel S, Satoi S, White SA, Hackert T, Nguyen TK, Yamamoto T, Nakamura T, Bachu V, Burns WR, Inoue Y, Takahashi Y, Ushida Y, Aslami ZV, Verbeke CS, Fariña A, He J, Wilmink JW, Messersmith W, Verheij J, Kaplan J, Schulick RD, Besselink MG, Del Chiaro M. Pathological Complete Response in Patients With Resected Pancreatic Adenocarcinoma After Preoperative Chemotherapy. JAMA Netw Open. 2024;7:e2417625. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 12] [Cited by in RCA: 18] [Article Influence: 18.0] [Reference Citation Analysis (0)] |
| 132. | Katz MHG, Shi Q, Meyers J, Herman JM, Chuong M, Wolpin BM, Ahmad S, Marsh R, Schwartz L, Behr S, Frankel WL, Collisson E, Leenstra J, Williams TM, Vaccaro G, Venook A, Meyerhardt JA, O'Reilly EM. Efficacy of Preoperative mFOLFIRINOX vs mFOLFIRINOX Plus Hypofractionated Radiotherapy for Borderline Resectable Adenocarcinoma of the Pancreas: The A021501 Phase 2 Randomized Clinical Trial. JAMA Oncol. 2022;8:1263-1270. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 95] [Cited by in RCA: 197] [Article Influence: 65.7] [Reference Citation Analysis (0)] |
| 133. | Franklin O, Sugawara T, Ross RB, Rodriguez Franco S, Colborn K, Karam S, Schulick RD, Del Chiaro M. Adjuvant Chemotherapy With or Without Radiotherapy for Resected Pancreatic Cancer After Multiagent Neoadjuvant Chemotherapy. Ann Surg Oncol. 2024;31:4966-4975. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 3] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
| 134. | Tang P, Zhang J, Zhou Q, Yi W, Wang H. Effect of Radiotherapy in Neoadjuvant Treatment of Borderline Resectable and Locally Advanced Pancreatic Cancer: A Systematic Review and Meta-analysis. Pancreas. 2025;54:e246-e254. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 135. | Dahan L, Williet N, Le Malicot K, Phelip JM, Desrame J, Bouché O, Petorin C, Malka D, Rebischung C, Aparicio T, Lecaille C, Rinaldi Y, Turpin A, Bignon AL, Bachet JB, Seitz JF, Lepage C, François E; PRODIGE 35 Investigators/Collaborators. Randomized Phase II Trial Evaluating Two Sequential Treatments in First Line of Metastatic Pancreatic Cancer: Results of the PANOPTIMOX-PRODIGE 35 Trial. J Clin Oncol. 2021;39:3242-3250. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 15] [Cited by in RCA: 49] [Article Influence: 12.3] [Reference Citation Analysis (0)] |
| 136. | Food and Drug Administration. FDA approves irinotecan liposome for first-line treatment of metastatic pancreatic adenocarcinoma. [cited September 22, 2025]. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-irinotecan-liposome-first-line-treatment-metastatic-pancreatic-adenocarcinoma. |
| 137. | Wainberg ZA, Melisi D, Macarulla T, Pazo Cid R, Chandana SR, De La Fouchardière C, Dean A, Kiss I, Lee WJ, Goetze TO, Van Cutsem E, Paulson AS, Bekaii-Saab T, Pant S, Hubner RA, Xiao Z, Chen H, Benzaghou F, O'Reilly EM. NALIRIFOX versus nab-paclitaxel and gemcitabine in treatment-naive patients with metastatic pancreatic ductal adenocarcinoma (NAPOLI 3): a randomised, open-label, phase 3 trial. Lancet. 2023;402:1272-1281. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 125] [Cited by in RCA: 214] [Article Influence: 107.0] [Reference Citation Analysis (0)] |
| 138. | Nichetti F, Rota S, Ambrosini P, Pircher C, Gusmaroli E, Droz Dit Busset M, Pusceddu S, Sposito C, Coppa J, Morano F, Pietrantonio F, Di Bartolomeo M, Mariani L, Mazzaferro V, de Braud F, Niger M. NALIRIFOX, FOLFIRINOX, and Gemcitabine With Nab-Paclitaxel as First-Line Chemotherapy for Metastatic Pancreatic Cancer: A Systematic Review and Meta-Analysis. JAMA Netw Open. 2024;7:e2350756. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 51] [Cited by in RCA: 62] [Article Influence: 62.0] [Reference Citation Analysis (0)] |
| 139. | Von Hoff DD, Ramanathan RK, Borad MJ, Laheru DA, Smith LS, Wood TE, Korn RL, Desai N, Trieu V, Iglesias JL, Zhang H, Soon-Shiong P, Shi T, Rajeshkumar NV, Maitra A, Hidalgo M. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J Clin Oncol. 2011;29:4548-4554. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 755] [Cited by in RCA: 887] [Article Influence: 63.4] [Reference Citation Analysis (2)] |
| 140. | Yang HK, Park MS, Choi M, Shin J, Lee SS, Jeong WK, Hwang SH, Choi SH. Systematic review and meta-analysis of diagnostic performance of CT imaging for assessing resectability of pancreatic ductal adenocarcinoma after neoadjuvant therapy: importance of CT criteria. Abdom Radiol (NY). 2021;46:5201-5217. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 11] [Article Influence: 2.8] [Reference Citation Analysis (0)] |
| 141. | de Jong TL, Koopman D, van der Worp CAJ, Stevens H, Vuijk FA, Vahrmeijer AL, Mieog JSD, de Groot JB, Meijssen MAC, Nieuwenhuijs VB, de Geus-Oei LF, Jager PL, Patijn GA. Added value of digital FDG-PET/CT in disease staging and restaging in patients with resectable or borderline resectable pancreatic cancer. Surg Oncol. 2023;47:101909. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 9] [Article Influence: 4.5] [Reference Citation Analysis (0)] |
| 142. | Wattenberg MM, Asch D, Yu S, O'Dwyer PJ, Domchek SM, Nathanson KL, Rosen MA, Beatty GL, Siegelman ES, Reiss KA. Platinum response characteristics of patients with pancreatic ductal adenocarcinoma and a germline BRCA1, BRCA2 or PALB2 mutation. Br J Cancer. 2020;122:333-339. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 124] [Cited by in RCA: 154] [Article Influence: 30.8] [Reference Citation Analysis (0)] |
| 143. | O'Reilly EM, Lee JW, Zalupski M, Capanu M, Park J, Golan T, Tahover E, Lowery MA, Chou JF, Sahai V, Brenner R, Kindler HL, Yu KH, Zervoudakis A, Vemuri S, Stadler ZK, Do RKG, Dhani N, Chen AP, Kelsen DP. Randomized, Multicenter, Phase II Trial of Gemcitabine and Cisplatin With or Without Veliparib in Patients With Pancreas Adenocarcinoma and a Germline BRCA/PALB2 Mutation. J Clin Oncol. 2020;38:1378-1388. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 252] [Cited by in RCA: 288] [Article Influence: 57.6] [Reference Citation Analysis (0)] |
| 144. | Erratum: Overall Survival Results From the POLO Trial: A Phase III Study of Active Maintenance Olaparib Versus Placebo for Germline BRCA-Mutated Metastatic Pancreatic Cancer. J Clin Oncol. 2024;42:2112. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 145. | Reiss KA, Mick R, O'Hara MH, Teitelbaum U, Karasic TB, Schneider C, Cowden S, Southwell T, Romeo J, Izgur N, Hannan ZM, Tondon R, Nathanson K, Vonderheide RH, Wattenberg MM, Beatty G, Domchek SM. Phase II Study of Maintenance Rucaparib in Patients With Platinum-Sensitive Advanced Pancreatic Cancer and a Pathogenic Germline or Somatic Variant in BRCA1, BRCA2, or PALB2. J Clin Oncol. 2021;39:2497-2505. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 43] [Cited by in RCA: 155] [Article Influence: 38.8] [Reference Citation Analysis (0)] |
| 146. | Li H, Liu ZY, Wu N, Chen YC, Cheng Q, Wang J. PARP inhibitor resistance: the underlying mechanisms and clinical implications. Mol Cancer. 2020;19:107. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 103] [Cited by in RCA: 337] [Article Influence: 67.4] [Reference Citation Analysis (0)] |
| 147. | Skoulidis F, Li BT, Dy GK, Price TJ, Falchook GS, Wolf J, Italiano A, Schuler M, Borghaei H, Barlesi F, Kato T, Curioni-Fontecedro A, Sacher A, Spira A, Ramalingam SS, Takahashi T, Besse B, Anderson A, Ang A, Tran Q, Mather O, Henary H, Ngarmchamnanrith G, Friberg G, Velcheti V, Govindan R. Sotorasib for Lung Cancers with KRAS p.G12C Mutation. N Engl J Med. 2021;384:2371-2381. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 941] [Cited by in RCA: 1194] [Article Influence: 298.5] [Reference Citation Analysis (0)] |
| 148. | Bekaii-Saab TS, Yaeger R, Spira AI, Pelster MS, Sabari JK, Hafez N, Barve M, Velastegui K, Yan X, Shetty A, Der-Torossian H, Pant S. Adagrasib in Advanced Solid Tumors Harboring a KRAS(G12C) Mutation. J Clin Oncol. 2023;41:4097-4106. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 27] [Cited by in RCA: 170] [Article Influence: 85.0] [Reference Citation Analysis (0)] |
| 149. | Hallin J, Bowcut V, Calinisan A, Briere DM, Hargis L, Engstrom LD, Laguer J, Medwid J, Vanderpool D, Lifset E, Trinh D, Hoffman N, Wang X, David Lawson J, Gunn RJ, Smith CR, Thomas NC, Martinson M, Bergstrom A, Sullivan F, Bouhana K, Winski S, He L, Fernandez-Banet J, Pavlicek A, Haling JR, Rahbaek L, Marx MA, Olson P, Christensen JG. Anti-tumor efficacy of a potent and selective non-covalent KRAS(G12D) inhibitor. Nat Med. 2022;28:2171-2182. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 300] [Article Influence: 100.0] [Reference Citation Analysis (0)] |
| 150. | NIH. Study of MRTX1133 in Patients With Advanced Solid Tumors Harboring a KRAS G12D Mutation. [cited September 22, 2025]. Available from: https://clinicaltrials.gov/ct2/show/NCT05737706. |
| 151. | Jiang J, Jiang L, Maldonato BJ, Wang Y, Holderfield M, Aronchik I, Winters IP, Salman Z, Blaj C, Menard M, Brodbeck J, Chen Z, Wei X, Rosen MJ, Gindin Y, Lee BJ, Evans JW, Chang S, Wang Z, Seamon KJ, Parsons D, Cregg J, Marquez A, Tomlinson ACA, Yano JK, Knox JE, Quintana E, Aguirre AJ, Arbour KC, Reed A, Gustafson WC, Gill AL, Koltun ES, Wildes D, Smith JAM, Wang Z, Singh M. Translational and Therapeutic Evaluation of RAS-GTP Inhibition by RMC-6236 in RAS-Driven Cancers. Cancer Discov. 2024;14:994-1017. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 13] [Cited by in RCA: 139] [Article Influence: 139.0] [Reference Citation Analysis (0)] |
| 152. | Arbour K, Punekar S, Garrido-laguna I, Hong D, Wolpin B, Pelster M, Barve M, Starodub A, Sommerhalder D, Chang S, Zhang Y, Salman Z, Wang X, Gustafson C, Spira A. 652O Preliminary clinical activity of RMC-6236, a first-in-class, RAS-selective, tri-complex RAS-MULTI(ON) inhibitor in patients with KRAS mutant pancreatic ductal adenocarcinoma (PDAC) and non-small cell lung cancer (NSCLC). Ann Oncol. 2023;34:S458. [DOI] [Full Text] |
| 153. | NIH. Phase 3 Study of Daraxonrasib (RMC-6236) in Patients With Previously Treated Metastatic Pancreatic Ductal Adenocarcinoma (PDAC) (RASolute 302). [cited September 22, 2025]. Available from: https://clinicaltrials.gov/ct2/show/NCT06625320. |
| 154. | Chung V, Spira A, Pavlick A, Sommerhalder D, Ma B, Hayreh V, de Jong J, Funt J, Kolitz S, Nair P, King P, Zhang J, Kim J, Yamamura A, Zeskind B, Hall B, Pant S. 1524P Preliminary phase I safety and activity of IMM-1-104, an orally dosed universal RAS inhibitor that drives deep cyclic inhibition of the MAPK pathway at MEK, in patients with advanced unresectable or metastatic solid tumors. Ann Oncol. 2024;35:S930-S931. [DOI] [Full Text] |
| 155. | Hillig RC, Sautier B, Schroeder J, Moosmayer D, Hilpmann A, Stegmann CM, Werbeck ND, Briem H, Boemer U, Weiske J, Badock V, Mastouri J, Petersen K, Siemeister G, Kahmann JD, Wegener D, Böhnke N, Eis K, Graham K, Wortmann L, von Nussbaum F, Bader B. Discovery of potent SOS1 inhibitors that block RAS activation via disruption of the RAS-SOS1 interaction. Proc Natl Acad Sci U S A. 2019;116:2551-2560. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 156] [Cited by in RCA: 299] [Article Influence: 49.8] [Reference Citation Analysis (0)] |
| 156. | Hofmann MH, Gmachl M, Ramharter J, Savarese F, Gerlach D, Marszalek JR, Sanderson MP, Kessler D, Trapani F, Arnhof H, Rumpel K, Botesteanu DA, Ettmayer P, Gerstberger T, Kofink C, Wunberg T, Zoephel A, Fu SC, Teh JL, Böttcher J, Pototschnig N, Schachinger F, Schipany K, Lieb S, Vellano CP, O'Connell JC, Mendes RL, Moll J, Petronczki M, Heffernan TP, Pearson M, McConnell DB, Kraut N. BI-3406, a Potent and Selective SOS1-KRAS Interaction Inhibitor, Is Effective in KRAS-Driven Cancers through Combined MEK Inhibition. Cancer Discov. 2021;11:142-157. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 98] [Cited by in RCA: 309] [Article Influence: 61.8] [Reference Citation Analysis (0)] |
| 157. | Punekar SR, Velcheti V, Neel BG, Wong KK. The current state of the art and future trends in RAS-targeted cancer therapies. Nat Rev Clin Oncol. 2022;19:637-655. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 329] [Reference Citation Analysis (0)] |
| 158. | Bery N, Miller A, Rabbitts T. A potent KRAS macromolecule degrader specifically targeting tumours with mutant KRAS. Nat Commun. 2020;11:3233. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 66] [Cited by in RCA: 89] [Article Influence: 17.8] [Reference Citation Analysis (0)] |
| 159. | Nagashima T, Inamura K, Nishizono Y, Suzuki A, Tanaka H, Yoshinari T, Yamanaka Y. 85 (PB075) - ASP3082, a First-in-class novel KRAS G12D degrader, exhibits remarkable anti-tumor activity in KRAS G12D mutated cancer models. 34th EORTC-NCI-AACR Symposium on Molecular Targets and Cancer Therapeutics; 2022 Oct 26-28; Barcelona, Spain. |
| 160. | Holderfield M, Lee BJ, Jiang J, Tomlinson A, Seamon KJ, Mira A, Patrucco E, Goodhart G, Dilly J, Gindin Y, Dinglasan N, Wang Y, Lai LP, Cai S, Jiang L, Nasholm N, Shifrin N, Blaj C, Shah H, Evans JW, Montazer N, Lai O, Shi J, Ahler E, Quintana E, Chang S, Salvador A, Marquez A, Cregg J, Liu Y, Milin A, Chen A, Ziv TB, Parsons D, Knox JE, Klomp JE, Roth J, Rees M, Ronan M, Cuevas-Navarro A, Hu F, Lito P, Santamaria D, Aguirre AJ, Waters AM, Der CJ, Ambrogio C, Wang Z, Gill AL, Koltun ES, Smith JAM, Wildes D, Singh M. Concurrent inhibition of oncogenic and wild-type RAS-GTP for cancer therapy. Nature. 2024;629:919-926. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 67] [Cited by in RCA: 156] [Article Influence: 156.0] [Reference Citation Analysis (0)] |
| 161. | Wasko UN, Jiang J, Dalton TC, Curiel-Garcia A, Edwards AC, Wang Y, Lee B, Orlen M, Tian S, Stalnecker CA, Drizyte-Miller K, Menard M, Dilly J, Sastra SA, Palermo CF, Hasselluhn MC, Decker-Farrell AR, Chang S, Jiang L, Wei X, Yang YC, Helland C, Courtney H, Gindin Y, Muonio K, Zhao R, Kemp SB, Clendenin C, Sor R, Vostrejs WP, Hibshman PS, Amparo AM, Hennessey C, Rees MG, Ronan MM, Roth JA, Brodbeck J, Tomassoni L, Bakir B, Socci ND, Herring LE, Barker NK, Wang J, Cleary JM, Wolpin BM, Chabot JA, Kluger MD, Manji GA, Tsai KY, Sekulic M, Lagana SM, Califano A, Quintana E, Wang Z, Smith JAM, Holderfield M, Wildes D, Lowe SW, Badgley MA, Aguirre AJ, Vonderheide RH, Stanger BZ, Baslan T, Der CJ, Singh M, Olive KP. Author Correction: Tumour-selective activity of RAS-GTP inhibition in pancreatic cancer. Nature. 2024;635:E12. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 162. | Kim DW, Schram AM, Hollebecque A, Nishino K, Macarulla T, Rha SY, Duruisseaux M, Liu SV, Al Hallak MN, Umemoto K, Wesseler C, Cleary JM, Springfeld C, Neuzillet C, Joe A, Jauhari S, Ford J, Goto K. The phase I/II eNRGy trial: Zenocutuzumab in patients with cancers harboring NRG1 gene fusions. Future Oncol. 2024;20:1057-1067. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 3] [Cited by in RCA: 30] [Article Influence: 30.0] [Reference Citation Analysis (0)] |
| 163. | Subbiah V, Lassen U, Élez E, Italiano A, Curigliano G, Javle M, de Braud F, Prager GW, Greil R, Stein A, Fasolo A, Schellens JHM, Wen PY, Viele K, Boran AD, Gasal E, Burgess P, Ilankumaran P, Wainberg ZA. Dabrafenib plus trametinib in patients with BRAF(V600E)-mutated biliary tract cancer (ROAR): a phase 2, open-label, single-arm, multicentre basket trial. Lancet Oncol. 2020;21:1234-1243. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 149] [Cited by in RCA: 353] [Article Influence: 70.6] [Reference Citation Analysis (1)] |
| 164. | Shen Z, Qiu B, Li L, Yang B, Li G. Targeted therapy of RET fusion-positive non-small cell lung cancer. Front Oncol. 2022;12:1033484. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 14] [Reference Citation Analysis (0)] |
| 165. | Subbiah V, Wolf J, Konda B, Kang H, Spira A, Weiss J, Takeda M, Ohe Y, Khan S, Ohashi K, Soldatenkova V, Szymczak S, Sullivan L, Wright J, Drilon A. Tumour-agnostic efficacy and safety of selpercatinib in patients with RET fusion-positive solid tumours other than lung or thyroid tumours (LIBRETTO-001): a phase 1/2, open-label, basket trial. Lancet Oncol. 2022;23:1261-1273. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 262] [Article Influence: 87.3] [Reference Citation Analysis (0)] |
| 166. | Meric-Bernstam F, Makker V, Oaknin A, Oh DY, Banerjee S, González-Martín A, Jung KH, Ługowska I, Manso L, Manzano A, Melichar B, Siena S, Stroyakovskiy D, Fielding A, Ma Y, Puvvada S, Shire N, Lee JY. Efficacy and Safety of Trastuzumab Deruxtecan in Patients With HER2-Expressing Solid Tumors: Primary Results From the DESTINY-PanTumor02 Phase II Trial. J Clin Oncol. 2024;42:47-58. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 112] [Cited by in RCA: 469] [Article Influence: 469.0] [Reference Citation Analysis (0)] |
| 167. | Hong DS, DuBois SG, Kummar S, Farago AF, Albert CM, Rohrberg KS, van Tilburg CM, Nagasubramanian R, Berlin JD, Federman N, Mascarenhas L, Geoerger B, Dowlati A, Pappo AS, Bielack S, Doz F, McDermott R, Patel JD, Schilder RJ, Tahara M, Pfister SM, Witt O, Ladanyi M, Rudzinski ER, Nanda S, Childs BH, Laetsch TW, Hyman DM, Drilon A. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol. 2020;21:531-540. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 335] [Cited by in RCA: 727] [Article Influence: 145.4] [Reference Citation Analysis (0)] |
| 168. | Doebele RC, Drilon A, Paz-Ares L, Siena S, Shaw AT, Farago AF, Blakely CM, Seto T, Cho BC, Tosi D, Besse B, Chawla SP, Bazhenova L, Krauss JC, Chae YK, Barve M, Garrido-Laguna I, Liu SV, Conkling P, John T, Fakih M, Sigal D, Loong HH, Buchschacher GL Jr, Garrido P, Nieva J, Steuer C, Overbeck TR, Bowles DW, Fox E, Riehl T, Chow-Maneval E, Simmons B, Cui N, Johnson A, Eng S, Wilson TR, Demetri GD; trial investigators. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol. 2020;21:271-282. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 574] [Cited by in RCA: 1184] [Article Influence: 197.3] [Reference Citation Analysis (0)] |
| 169. | Drilon A, Camidge DR, Lin JJ, Kim SW, Solomon BJ, Dziadziuszko R, Besse B, Goto K, de Langen AJ, Wolf J, Lee KH, Popat S, Springfeld C, Nagasaka M, Felip E, Yang N, Velcheti V, Lu S, Kao S, Dooms C, Krebs MG, Yao W, Beg MS, Hu X, Moro-Sibilot D, Cheema P, Stopatschinskaja S, Mehta M, Trone D, Graber A, Sims G, Yuan Y, Cho BC; TRIDENT-1 Investigators. Repotrectinib in ROS1 Fusion-Positive Non-Small-Cell Lung Cancer. N Engl J Med. 2024;390:118-131. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 109] [Cited by in RCA: 109] [Article Influence: 109.0] [Reference Citation Analysis (0)] |
| 170. | Cereda V, D'Andrea MR. Pancreatic cancer: failures and hopes-a review of new promising treatment approaches. Explor Target Antitumor Ther. 2025;6:1002299. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 171. | Gupta N, Huang TT, Horibata S, Lee JM. Cell cycle checkpoints and beyond: Exploiting the ATR/CHK1/WEE1 pathway for the treatment of PARP inhibitor-resistant cancer. Pharmacol Res. 2022;178:106162. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 45] [Cited by in RCA: 74] [Article Influence: 24.7] [Reference Citation Analysis (0)] |
| 172. | Gorecki L, Andrs M, Korabecny J. Clinical Candidates Targeting the ATR-CHK1-WEE1 Axis in Cancer. Cancers (Basel). 2021;13:795. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 38] [Cited by in RCA: 58] [Article Influence: 14.5] [Reference Citation Analysis (1)] |
| 173. | Jones R, Plummer R, Moreno V, Carter L, Roda D, Garralda E, Kristeleit R, Sarker D, Arkenau T, Roxburgh P, Walter HS, Blagden S, Anthoney A, Klencke BJ, Kowalski MM, Banerji U. A Phase I/II Trial of Oral SRA737 (a Chk1 Inhibitor) Given in Combination with Low-Dose Gemcitabine in Patients with Advanced Cancer. Clin Cancer Res. 2023;29:331-340. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 8] [Cited by in RCA: 30] [Article Influence: 15.0] [Reference Citation Analysis (0)] |
| 174. | Cleary JM, Wolpin BM, Dougan SK, Raghavan S, Singh H, Huffman B, Sethi NS, Nowak JA, Shapiro GI, Aguirre AJ, D'Andrea AD. Opportunities for Utilization of DNA Repair Inhibitors in Homologous Recombination Repair-Deficient and Proficient Pancreatic Adenocarcinoma. Clin Cancer Res. 2021;27:6622-6637. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 12] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
| 175. | Cuneo KC, Morgan MA, Sahai V, Schipper MJ, Parsels LA, Parsels JD, Devasia T, Al-Hawaray M, Cho CS, Nathan H, Maybaum J, Zalupski MM, Lawrence TS. Dose Escalation Trial of the Wee1 Inhibitor Adavosertib (AZD1775) in Combination With Gemcitabine and Radiation for Patients With Locally Advanced Pancreatic Cancer. J Clin Oncol. 2019;37:2643-2650. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 79] [Cited by in RCA: 150] [Article Influence: 25.0] [Reference Citation Analysis (0)] |
| 176. | Stein L, Murugesan K, Reeser JW, Risch Z, Wing MR, Paruchuri A, Samorodnitsky E, Hoskins EL, Dao T, Smith A, Le D, Babcook MA, Chang YS, Avenarius MR, Imam M, Freud AG, Roychowdhury S. FGFR2-fusions define a clinically actionable molecular subset of pancreatic cancer. NPJ Precis Oncol. 2024;8:207. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 6] [Reference Citation Analysis (0)] |
| 177. | Subbiah V, Sahai V, Maglic D, Bruderek K, Touré BB, Zhao S, Valverde R, O'Hearn PJ, Moustakas DT, Schönherr H, Gerami-Moayed N, Taylor AM, Hudson BM, Houde DJ, Pal D, Foster L, Gunaydin H, Ayaz P, Sharon DA, Goyal L, Schram AM, Kamath S, Sherwin CA, Schmidt-Kittler O, Jen KY, Ricard F, Wolf BB, Shaw DE, Bergstrom DA, Watters J, Casaletto JB. RLY-4008, the First Highly Selective FGFR2 Inhibitor with Activity across FGFR2 Alterations and Resistance Mutations. Cancer Discov. 2023;13:2012-2031. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 38] [Cited by in RCA: 81] [Article Influence: 40.5] [Reference Citation Analysis (0)] |
| 178. | Rodon J, Prenen H, Sacher A, Villalona-Calero M, Penel N, El Helali A, Rottey S, Yamamoto N, Ghiringhelli F, Goebeler ME, Doi T, Postel-Vinay S, Lin CC, Liu C, Chuang CH, Keyvanjah K, Eggert T, O'Neil BH. First-in-human study of AMG 193, an MTA-cooperative PRMT5 inhibitor, in patients with MTAP-deleted solid tumors: results from phase I dose exploration. Ann Oncol. 2024;35:1138-1147. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 36] [Reference Citation Analysis (0)] |
| 179. | Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, Kemberling H, Wilt C, Luber BS, Wong F, Azad NS, Rucki AA, Laheru D, Donehower R, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Greten TF, Duffy AG, Ciombor KK, Eyring AD, Lam BH, Joe A, Kang SP, Holdhoff M, Danilova L, Cope L, Meyer C, Zhou S, Goldberg RM, Armstrong DK, Bever KM, Fader AN, Taube J, Housseau F, Spetzler D, Xiao N, Pardoll DM, Papadopoulos N, Kinzler KW, Eshleman JR, Vogelstein B, Anders RA, Diaz LA Jr. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409-413. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3799] [Cited by in RCA: 5036] [Article Influence: 629.5] [Reference Citation Analysis (0)] |
| 180. | Marabelle A, Le DT, Ascierto PA, Di Giacomo AM, De Jesus-Acosta A, Delord JP, Geva R, Gottfried M, Penel N, Hansen AR, Piha-Paul SA, Doi T, Gao B, Chung HC, Lopez-Martin J, Bang YJ, Frommer RS, Shah M, Ghori R, Joe AK, Pruitt SK, Diaz LA Jr. Efficacy of Pembrolizumab in Patients With Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J Clin Oncol. 2020;38:1-10. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2120] [Cited by in RCA: 2102] [Article Influence: 420.4] [Reference Citation Analysis (0)] |
| 181. | Le DT, Kim TW, Van Cutsem E, Geva R, Jäger D, Hara H, Burge M, O'Neil B, Kavan P, Yoshino T, Guimbaud R, Taniguchi H, Elez E, Al-Batran SE, Boland PM, Crocenzi T, Atreya CE, Cui Y, Dai T, Marinello P, Diaz LA Jr, André T. Phase II Open-Label Study of Pembrolizumab in Treatment-Refractory, Microsatellite Instability-High/Mismatch Repair-Deficient Metastatic Colorectal Cancer: KEYNOTE-164. J Clin Oncol. 2020;38:11-19. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 346] [Cited by in RCA: 724] [Article Influence: 120.7] [Reference Citation Analysis (0)] |
| 182. | Geoerger B, Kang HJ, Yalon-Oren M, Marshall LV, Vezina C, Pappo A, Laetsch TW, Petrilli AS, Ebinger M, Toporski J, Glade-Bender J, Nicholls W, Fox E, DuBois SG, Macy ME, Cohn SL, Pathiraja K, Diede SJ, Ebbinghaus S, Pinto N. Pembrolizumab in paediatric patients with advanced melanoma or a PD-L1-positive, advanced, relapsed, or refractory solid tumour or lymphoma (KEYNOTE-051): interim analysis of an open-label, single-arm, phase 1-2 trial. Lancet Oncol. 2020;21:121-133. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 109] [Cited by in RCA: 209] [Article Influence: 34.8] [Reference Citation Analysis (0)] |
| 183. | Coston T, Desai A, Babiker H, Sonbol MB, Chakrabarti S, Mahipal A, McWilliams R, Ma WW, Bekaii-Saab TS, Stauffer J, Starr JS. Efficacy of Immune Checkpoint Inhibition and Cytotoxic Chemotherapy in Mismatch Repair-Deficient and Microsatellite Instability-High Pancreatic Cancer: Mayo Clinic Experience. JCO Precis Oncol. 2023;7:e2200706. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 17] [Cited by in RCA: 23] [Article Influence: 11.5] [Reference Citation Analysis (0)] |
| 184. | Taïeb J, Sayah L, Heinrich K, Kunzmann V, Boileve A, Cirkel G, Lonardi S, Chibaudel B, Turpin A, Beller T, Hautefeuille V, Vivaldi C, Mazard T, Bauguion L, Niger M, Prager GW, Coutzac C, Benedikt Westphalen C, Auclin E, Pilla L. Efficacy of immune checkpoint inhibitors in microsatellite unstable/mismatch repair-deficient advanced pancreatic adenocarcinoma: an AGEO European Cohort. Eur J Cancer. 2023;188:90-97. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 47] [Article Influence: 23.5] [Reference Citation Analysis (0)] |
| 185. | Overman MJ, Melhem R, Blum-Murphy MA, Ramos C, Petrosyan L, Li J, Perer JK, Zou H, Wang M, Wright HM. A phase I, first-in-human, open-label, dose escalation and expansion study of PT886 in adult patients with advanced gastric, gastroesophageal junction, and pancreatic adenocarcinomas. J Clin Oncol. 2023;41:TPS765-TPS765. [RCA] [DOI] [Full Text] [Reference Citation Analysis (0)] |
| 186. | Dean A, Gill S, McGregor M, Broadbridge V, Järveläinen HA, Price T. Dual αV-integrin and neuropilin-1 targeting peptide CEND-1 plus nab-paclitaxel and gemcitabine for the treatment of metastatic pancreatic ductal adenocarcinoma: a first-in-human, open-label, multicentre, phase 1 study. Lancet Gastroenterol Hepatol. 2022;7:943-951. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 18] [Cited by in RCA: 47] [Article Influence: 15.7] [Reference Citation Analysis (0)] |
| 187. | Hurtado de Mendoza T, Mose ES, Botta GP, Braun GB, Kotamraju VR, French RP, Suzuki K, Miyamura N, Teesalu T, Ruoslahti E, Lowy AM, Sugahara KN. Tumor-penetrating therapy for β5 integrin-rich pancreas cancer. Nat Commun. 2021;12:1541. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 35] [Cited by in RCA: 54] [Article Influence: 13.5] [Reference Citation Analysis (0)] |
| 188. | Kasi A, Jarvelainen H, Al-Rajabi RMT, Saeed A, Phadnis MA, Chidharla A, Schmitt T, Kumer S, Al-Kasspooles MM, Ashcraft J, Martin B, Luka S, Olyaee M, Rastogi A, Weir SJ, Saha S, Dandawate P, Madan R, Sun W, Baranda JC. Phase Ib/IIa trial of CEND‐1 in combination with neoadjuvant FOLFIRINOX-based therapies in pancreatic, colorectal, and appendiceal cancers (CENDIFOX). J Clin Oncol. 2022;40:TPS4195. [RCA] [DOI] [Full Text] [Reference Citation Analysis (0)] |
| 189. | Pant M, Furgan M, Abdul-Karim RM, Chung V, Devoe CE, Johnson ML, Leal AD, Park H, Wainberg ZA, Welkowsky E, Haqq CM, O’Reilly EM, Weekes CD. First-in-human phase 1 trial of ELI-002 immunotherapy as treatment for subjects with Kirsten rat sarcoma (KRAS)-mutated pancreatic ductal adenocarcinoma and other solid tumors. J Clin Oncol. 2022;40:TPS2701. [RCA] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 2] [Article Influence: 0.7] [Reference Citation Analysis (0)] |
| 190. | O'Reilly EM, Wainberg ZA, Weekes CD, Furqan M, Kasi PM, Devoe CE, Leal AD, Chung V, Perry J, Seenappa L, McNeil L, Welkowsky E, DeMuth P, Haqq CM, Pant S. AMPLIFY-201, a first-in-human safety and efficacy trial of adjuvant ELI-002 2P immunotherapy for patients with high-relapse risk with KRAS G12D- or G12R-mutated pancreatic and colorectal cancer. J Clin Oncol. 2023;41:2528. [DOI] [Full Text] |
| 191. | Surana R, LeBleu VS, Lee JJ, Smaglo BG, Zhao D, Lee MS, Wolff RA, Overman MJ, Mendt MC, McAndrews KM, Yang S, Rezvani K, Kalluri R, Maitra A, Shpall EJ, Pant S. Phase I study of mesenchymal stem cell (MSC)-derived exosomes with KRASG12D siRNA in patients with metastatic pancreatic cancer harboring a KRASG12D mutation. J Clin Oncol. 2022;40:TPS633. [RCA] [DOI] [Full Text] [Reference Citation Analysis (0)] |
| 192. | Heumann T, Judkins C, Li K, Lim SJ, Hoare J, Parkinson R, Cao H, Zhang T, Gai J, Celiker B, Zhu Q, McPhaul T, Durham J, Purtell K, Klein R, Laheru D, De Jesus-Acosta A, Le DT, Narang A, Anders R, Burkhart R, Burns W, Soares K, Wolfgang C, Thompson E, Jaffee E, Wang H, He J, Zheng L. A platform trial of neoadjuvant and adjuvant antitumor vaccination alone or in combination with PD-1 antagonist and CD137 agonist antibodies in patients with resectable pancreatic adenocarcinoma. Nat Commun. 2023;14:3650. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 18] [Cited by in RCA: 54] [Article Influence: 27.0] [Reference Citation Analysis (0)] |
| 193. | Rojas LA, Sethna Z, Soares KC, Olcese C, Pang N, Patterson E, Lihm J, Ceglia N, Guasp P, Chu A, Yu R, Chandra AK, Waters T, Ruan J, Amisaki M, Zebboudj A, Odgerel Z, Payne G, Derhovanessian E, Müller F, Rhee I, Yadav M, Dobrin A, Sadelain M, Łuksza M, Cohen N, Tang L, Basturk O, Gönen M, Katz S, Do RK, Epstein AS, Momtaz P, Park W, Sugarman R, Varghese AM, Won E, Desai A, Wei AC, D'Angelica MI, Kingham TP, Mellman I, Merghoub T, Wolchok JD, Sahin U, Türeci Ö, Greenbaum BD, Jarnagin WR, Drebin J, O'Reilly EM, Balachandran VP. Personalized RNA neoantigen vaccines stimulate T cells in pancreatic cancer. Nature. 2023;618:144-150. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 827] [Cited by in RCA: 760] [Article Influence: 380.0] [Reference Citation Analysis (0)] |
| 194. | Musher BL, Smaglo BG, Abidi W, Othman M, Patel K, Jawaid S, Jing J, Brisco A, Wenthe J, Eriksson E, Ullenhag GJ, Sandin L, Grilley B, Leja-jarblad J, Hilsenbeck SG, Brenner MK, Rowinsky EK, Loskog ASI. A phase I/II study of LOAd703, a TMZ-CD40L/4-1BBL-armed oncolytic adenovirus, combined with nab-paclitaxel and gemcitabine in advanced pancreatic cancer. J Clin Oncol. 2022;40:4138. [RCA] [DOI] [Full Text] [Cited by in Crossref: 3] [Cited by in RCA: 4] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
| 195. | Mahalingam D, Chen S, Xie P, Loghmani H, Heineman T, Kalyan A, Kircher S, Helenowski IB, Mi X, Maurer V, Coffey M, Mulcahy M, Benson A, Zhang B. Combination of pembrolizumab and pelareorep promotes anti-tumour immunity in advanced pancreatic adenocarcinoma (PDAC). Br J Cancer. 2023;129:782-790. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3] [Cited by in RCA: 12] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
| 196. | Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 2021;11:69. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 326] [Cited by in RCA: 1680] [Article Influence: 420.0] [Reference Citation Analysis (0)] |
| 197. | Czaplicka A, Lachota M, Pączek L, Zagożdżon R, Kaleta B. Chimeric Antigen Receptor T Cell Therapy for Pancreatic Cancer: A Review of Current Evidence. Cells. 2024;13:101. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 6] [Cited by in RCA: 20] [Article Influence: 20.0] [Reference Citation Analysis (0)] |
| 198. | Liu Y, Guo Y, Wu Z, Feng K, Tong C, Wang Y, Dai H, Shi F, Yang Q, Han W. Anti-EGFR chimeric antigen receptor-modified T cells in metastatic pancreatic carcinoma: A phase I clinical trial. Cytotherapy. 2020;22:573-580. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 66] [Cited by in RCA: 134] [Article Influence: 26.8] [Reference Citation Analysis (0)] |
| 199. | Cutmore LC, Brown NF, Raj D, Chauduri S, Wang P, Maher J, Wang Y, Lemoine NR, Marshall JF. Pancreatic Cancer UK Grand Challenge: Developments and challenges for effective CAR T cell therapy for pancreatic ductal adenocarcinoma. Pancreatology. 2020;20:394-408. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 7] [Cited by in RCA: 12] [Article Influence: 2.4] [Reference Citation Analysis (0)] |
| 200. | Haas AR, Tanyi JL, O'Hara MH, Gladney WL, Lacey SF, Torigian DA, Soulen MC, Tian L, McGarvey M, Nelson AM, Farabaugh CS, Moon E, Levine BL, Melenhorst JJ, Plesa G, June CH, Albelda SM, Beatty GL. Phase I Study of Lentiviral-Transduced Chimeric Antigen Receptor-Modified T Cells Recognizing Mesothelin in Advanced Solid Cancers. Mol Ther. 2019;27:1919-1929. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 121] [Cited by in RCA: 277] [Article Influence: 46.2] [Reference Citation Analysis (0)] |
| 201. | Botta GP, Becerra CR, Jin Z, Kim DW, Zhao D, Lenz H, Ma H, Ween A, Acha P, Li Z, Yoon HH. Multicenter phase Ib trial in the U.S. of salvage CT041 CLDN18.2-specific chimeric antigen receptor T-cell therapy for patients with advanced gastric and pancreatic adenocarcinoma. J Clin Oncol. 2022;40:2538. [RCA] [DOI] [Full Text] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
| 202. | Qi C, Gong J, Li J, Liu D, Qin Y, Ge S, Zhang M, Peng Z, Zhou J, Cao Y, Zhang X, Lu Z, Lu M, Yuan J, Wang Z, Wang Y, Peng X, Gao H, Liu Z, Wang H, Yuan D, Xiao J, Ma H, Wang W, Li Z, Shen L. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat Med. 2022;28:1189-1198. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 317] [Cited by in RCA: 404] [Article Influence: 134.7] [Reference Citation Analysis (36)] |
| 203. | Wang Y, Chen M, Wu Z, Tong C, Dai H, Guo Y, Liu Y, Huang J, Lv H, Luo C, Feng KC, Yang QM, Li XL, Han W. CD133-directed CAR T cells for advanced metastasis malignancies: A phase I trial. Oncoimmunology. 2018;7:e1440169. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 130] [Cited by in RCA: 257] [Article Influence: 36.7] [Reference Citation Analysis (0)] |
| 204. | Chen T, Wang M, Chen Y, Liu Y. Current challenges and therapeutic advances of CAR-T cell therapy for solid tumors. Cancer Cell Int. 2024;24:133. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 6] [Cited by in RCA: 57] [Article Influence: 57.0] [Reference Citation Analysis (0)] |
| 205. | Posey AD Jr, Schwab RD, Boesteanu AC, Steentoft C, Mandel U, Engels B, Stone JD, Madsen TD, Schreiber K, Haines KM, Cogdill AP, Chen TJ, Song D, Scholler J, Kranz DM, Feldman MD, Young R, Keith B, Schreiber H, Clausen H, Johnson LA, June CH. Engineered CAR T Cells Targeting the Cancer-Associated Tn-Glycoform of the Membrane Mucin MUC1 Control Adenocarcinoma. Immunity. 2016;44:1444-1454. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 454] [Cited by in RCA: 463] [Article Influence: 51.4] [Reference Citation Analysis (0)] |
| 206. | Henry J, Oh D, Eskew J, Baranda J, Rodriguez Rivera II, Dumbrava E, Cohen E, Belani R, Mccaigue J, Shedlock D, Coronella J, Martin C, Namini H, Murphy A, Ostertag E. 728 Phase 1 study of P-MUC1C-ALLO1 allogeneic CAR-T cells in patients with epithelial-derived cancers. J Immunother Cancer. 2022;10:A761. [DOI] [Full Text] |
| 207. | Gutierrez R, Shah PD, Hamid O, Garfall AL, Posey A, Bishop MR, Blumenschein GR, Johnson ML, Lee S, Luke JJ, Morgensztern D, Fountaine TJ, Dryer-minnerly R, Najmi S, Hufner P, Chagin K. Phase I experience with first in class TnMUC1 targeted chimeric antigen receptor T-cells in patients with advanced TnMUC1 positive solid tumors. J Clin Oncol. 2021;39:e14513. [RCA] [DOI] [Full Text] [Cited by in Crossref: 5] [Cited by in RCA: 5] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
| 208. | Farhangnia P, Khorramdelazad H, Nickho H, Delbandi AA. Current and future immunotherapeutic approaches in pancreatic cancer treatment. J Hematol Oncol. 2024;17:40. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 3] [Cited by in RCA: 93] [Article Influence: 93.0] [Reference Citation Analysis (0)] |
| 209. | Baulu E, Gardet C, Chuvin N, Depil S. TCR-engineered T cell therapy in solid tumors: State of the art and perspectives. Sci Adv. 2023;9:eadf3700. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 4] [Cited by in RCA: 232] [Article Influence: 116.0] [Reference Citation Analysis (0)] |
| 210. | Chiorean EG, Chapuis A, Coveler AL, Yeung CC, Gooley T, Zhen DB, King GT, Hannan LM, Cohen SA, Safyan RA, Germani A, Ra S, Casserd J, Schmitt T, Greenberg PD. Phase I study of autologous transgenic T cells expressing high affinity mesothelin-specific T-cell receptor (TCR; FH-TCR TMSLN) in patients with metastatic pancreatic ductal adenocarcinoma (mPDA). J Clin Oncol. 2023;41:TPS779-TPS779. [RCA] [DOI] [Full Text] [Reference Citation Analysis (0)] |
| 211. | Miller RA, Luke JJ, Hu S, Mahabhashyam S, Jones WB, Marron T, Merchan JR, Hughes BGM, Willingham SB. Anti-CD73 antibody activates human B cells, enhances humoral responses and induces redistribution of B cells in patients with cancer. J Immunother Cancer. 2022;10:e005802. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 18] [Reference Citation Analysis (0)] |
| 212. | Bendell J, LoRusso P, Overman M, Noonan AM, Kim DW, Strickler JH, Kim SW, Clarke S, George TJ, Grimison PS, Barve M, Amin M, Desai J, Wise-Draper T, Eck S, Jiang Y, Khan AA, Wu Y, Martin P, Cooper ZA, Elgeioushi N, Mueller N, Kumar R, Patel SP. First-in-human study of oleclumab, a potent, selective anti-CD73 monoclonal antibody, alone or in combination with durvalumab in patients with advanced solid tumors. Cancer Immunol Immunother. 2023;72:2443-2458. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 33] [Cited by in RCA: 63] [Article Influence: 31.5] [Reference Citation Analysis (0)] |
| 213. | Farhangnia P, Ghomi SM, Akbarpour M, Delbandi AA. Bispecific antibodies targeting CTLA-4: game-changer troopers in cancer immunotherapy. Front Immunol. 2023;14:1155778. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 6] [Cited by in RCA: 21] [Article Influence: 10.5] [Reference Citation Analysis (0)] |
| 214. | Long AW, Xu H, Santich BH, Guo H, Hoseini SS, de Stanchina E, Cheung NV. Heterodimerization of T cell engaging bispecific antibodies to enhance specificity against pancreatic ductal adenocarcinoma. J Hematol Oncol. 2024;17:20. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3] [Cited by in RCA: 8] [Article Influence: 8.0] [Reference Citation Analysis (0)] |
| 215. | Segal NH, Melero I, Moreno V, Steeghs N, Marabelle A, Rohrberg K, Rodriguez-Ruiz ME, Eder JP, Eng C, Manji GA, Waterkamp D, Leutgeb B, Bouseida S, Flinn N, Das Thakur M, Elze MC, Koeppen H, Jamois C, Martin-Facklam M, Lieu CH, Calvo E, Paz-Ares L, Tabernero J, Argilés G. CEA-CD3 bispecific antibody cibisatamab with or without atezolizumab in patients with CEA-positive solid tumours: results of two multi-institutional Phase 1 trials. Nat Commun. 2024;15:4091. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 7] [Cited by in RCA: 21] [Article Influence: 21.0] [Reference Citation Analysis (0)] |
| 216. | Schram AM, Odintsov I, Espinosa-Cotton M, Khodos I, Sisso WJ, Mattar MS, Lui AJW, Vojnic M, Shameem SH, Chauhan T, Torrisi J, Ford J, O'Connor MN, Geuijen CAW, Schackmann RCJ, Lammerts van Bueren JJ, Wasserman E, de Stanchina E, O'Reilly EM, Ladanyi M, Drilon A, Somwar R. Zenocutuzumab, a HER2xHER3 Bispecific Antibody, Is Effective Therapy for Tumors Driven by NRG1 Gene Rearrangements. Cancer Discov. 2022;12:1233-1247. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 14] [Cited by in RCA: 126] [Article Influence: 42.0] [Reference Citation Analysis (0)] |
| 217. | Thakur A, Ung J, Tomaszewski EN, Schienschang A, LaBrie TM, Schalk DL, Lum LG. Priming of pancreatic cancer cells with bispecific antibody armed activated T cells sensitizes tumors for enhanced chemoresponsiveness. Oncoimmunology. 2021;10:1930883. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 8] [Cited by in RCA: 12] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
| 218. | Oberg HH, Peipp M, Kellner C, Sebens S, Krause S, Petrick D, Adam-Klages S, Röcken C, Becker T, Vogel I, Weisner D, Freitag-Wolf S, Gramatzki M, Kabelitz D, Wesch D. Novel bispecific antibodies increase γδ T-cell cytotoxicity against pancreatic cancer cells. Cancer Res. 2014;74:1349-1360. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 92] [Cited by in RCA: 128] [Article Influence: 11.6] [Reference Citation Analysis (0)] |
| 219. | Akce M, Hu-lieskovan S, Reilley M, Strauss JF, Specht JM, Stein MN, Wang JS, Choe JH, Leidner R, Davar D, Falchook GS, Pant S, Cohen EE, Wilky BA, Thompson B, Clynes R, Li L, Mcgovern P, Liebowitz DN. A phase 1 multiple-ascending dose study to evaluate the safety and tolerability of XmAb23104 (PD-1 x ICOS) in subjects with selected advanced solid tumors (DUET-3). J Clin Oncol. 2022;40:2604. [RCA] [DOI] [Full Text] [Reference Citation Analysis (0)] |
| 220. | Gang J, Guo S, Zhang Y, Ma Y, Guo X, Zhou X, Yu Q. A phase II study of KN046 monotherapy as 2nd line and above treatment for unresectable locally advanced or metastatic pancreatic ductal adenocarcinoma (PDAC). J Clin Oncol. 2022;40:e16305. [RCA] [DOI] [Full Text] [Reference Citation Analysis (0)] |
| 221. | Sokol ES, Pavlick D, Khiabanian H, Frampton GM, Ross JS, Gregg JP, Lara PN, Oesterreich S, Agarwal N, Necchi A, Miller VA, Alexander B, Ali SM, Ganesan S, Chung JH. Pan-Cancer Analysis of BRCA1 and BRCA2 Genomic Alterations and Their Association With Genomic Instability as Measured by Genome-Wide Loss of Heterozygosity. JCO Precis Oncol. 2020;4:442-465. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 129] [Cited by in RCA: 126] [Article Influence: 25.2] [Reference Citation Analysis (0)] |
| 222. | Terrero G, Datta J, Dennison J, Sussman DA, Lohse I, Merchant NB, Hosein PJ. Ipilimumab/Nivolumab Therapy in Patients With Metastatic Pancreatic or Biliary Cancer With Homologous Recombination Deficiency Pathogenic Germline Variants. JAMA Oncol. 2022;8:1-3. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 45] [Article Influence: 15.0] [Reference Citation Analysis (0)] |
| 223. | Callahan M, Amin A, Kaye FJ, Morse MA, Taylor MH, Peltola KJ, Sharma P, O'Reilly EM, Meadows Shropshire S, O'Brien S, Tschaika M, Le DT. Nivolumab monotherapy or combination with ipilimumab with or without cobimetinib in previously treated patients with pancreatic adenocarcinoma (CheckMate 032). J Immunother Cancer. 2024;12:e007883. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 8] [Article Influence: 8.0] [Reference Citation Analysis (0)] |
| 224. | Reiss KA, Mick R, Teitelbaum U, O'Hara M, Schneider C, Massa R, Karasic T, Tondon R, Onyiah C, Gosselin MK, Donze A, Domchek SM, Vonderheide RH. Niraparib plus nivolumab or niraparib plus ipilimumab in patients with platinum-sensitive advanced pancreatic cancer: a randomised, phase 1b/2 trial. Lancet Oncol. 2022;23:1009-1020. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 35] [Cited by in RCA: 95] [Article Influence: 31.7] [Reference Citation Analysis (0)] |
| 225. | Park W, O'connor C, Chou JF, Schwartz C, Varghese AM, Larsen M, Balogun F, Brenner R, Yu KH, Diguglielmo E, Umeda S, Karnoub E, Keane F, Zhang H, Joshi SS, Riaz N, Kelsen DP, Capanu M, Iacobuzio-donahue CA, O'reilly EM. Phase 2 trial of pembrolizumab and olaparib (POLAR) maintenance for patients (pts) with metastatic pancreatic cancer (mPDAC): Two cohorts B non-core homologous recombination deficiency (HRD) and C exceptional response to platinum-therapy. J Clin Oncol. 2023;41:4140. [DOI] [Full Text] |
| 226. | Zhu H, Wei M, Xu J, Hua J, Liang C, Meng Q, Zhang Y, Liu J, Zhang B, Yu X, Shi S. PARP inhibitors in pancreatic cancer: molecular mechanisms and clinical applications. Mol Cancer. 2020;19:49. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 186] [Cited by in RCA: 186] [Article Influence: 37.2] [Reference Citation Analysis (0)] |
| 227. | Espona-Fiedler M, Patthey C, Lindblad S, Sarró I, Öhlund D. Overcoming therapy resistance in pancreatic cancer: New insights and future directions. Biochem Pharmacol. 2024;229:116492. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 14] [Article Influence: 14.0] [Reference Citation Analysis (0)] |
| 228. | Brown TJ, Reiss KA. PARP Inhibitors in Pancreatic Cancer. Cancer J. 2021;27:465-475. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 13] [Cited by in RCA: 35] [Article Influence: 8.8] [Reference Citation Analysis (0)] |
| 229. | Ho WJ, Jaffee EM, Zheng L. The tumour microenvironment in pancreatic cancer - clinical challenges and opportunities. Nat Rev Clin Oncol. 2020;17:527-540. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 854] [Cited by in RCA: 847] [Article Influence: 169.4] [Reference Citation Analysis (0)] |
| 230. | Kocher HM; BCI-STARPAC2 team; BPTB team; Precision-Panc team, Sasieni P, Corrie P, McNamara MG, Sarker D, Froeling FEM, Christie A, Gillmore R, Khan K, Propper D. Study protocol: multi-centre, randomised controlled clinical trial exploring stromal targeting in locally advanced pancreatic cancer; STARPAC2. BMC Cancer. 2025;25:106. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 5] [Article Influence: 5.0] [Reference Citation Analysis (0)] |
| 231. | Huang C, Shen Y, Galgano SJ, Goenka AH, Hecht EM, Kambadakone A, Wang ZJ, Chu LC. Advancements in early detection of pancreatic cancer: the role of artificial intelligence and novel imaging techniques. Abdom Radiol (NY). 2025;50:1731-1743. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2] [Cited by in RCA: 3] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
| 232. | Cao K, Xia Y, Yao J, Han X, Lambert L, Zhang T, Tang W, Jin G, Jiang H, Fang X, Nogues I, Li X, Guo W, Wang Y, Fang W, Qiu M, Hou Y, Kovarnik T, Vocka M, Lu Y, Chen Y, Chen X, Liu Z, Zhou J, Xie C, Zhang R, Lu H, Hager GD, Yuille AL, Lu L, Shao C, Shi Y, Zhang Q, Liang T, Zhang L, Lu J. Large-scale pancreatic cancer detection via non-contrast CT and deep learning. Nat Med. 2023;29:3033-3043. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 118] [Article Influence: 59.0] [Reference Citation Analysis (0)] |
| 233. | Si K, Xue Y, Yu X, Zhu X, Li Q, Gong W, Liang T, Duan S. Fully end-to-end deep-learning-based diagnosis of pancreatic tumors. Theranostics. 2021;11:1982-1990. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 19] [Cited by in RCA: 58] [Article Influence: 14.5] [Reference Citation Analysis (0)] |
| 234. | Ma H, Liu ZX, Zhang JJ, Wu FT, Xu CF, Shen Z, Yu CH, Li YM. Construction of a convolutional neural network classifier developed by computed tomography images for pancreatic cancer diagnosis. World J Gastroenterol. 2020;26:5156-5168. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in CrossRef: 56] [Cited by in RCA: 37] [Article Influence: 7.4] [Reference Citation Analysis (1)] |
| 235. | Marya NB, Powers PD, Chari ST, Gleeson FC, Leggett CL, Abu Dayyeh BK, Chandrasekhara V, Iyer PG, Majumder S, Pearson RK, Petersen BT, Rajan E, Sawas T, Storm AC, Vege SS, Chen S, Long Z, Hough DM, Mara K, Levy MJ. Utilisation of artificial intelligence for the development of an EUS-convolutional neural network model trained to enhance the diagnosis of autoimmune pancreatitis. Gut. 2021;70:1335-1344. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 44] [Cited by in RCA: 96] [Article Influence: 24.0] [Reference Citation Analysis (1)] |
| 236. | Zhao G, Chen X, Zhu M, Liu Y, Wang Y. Exploring the application and future outlook of Artificial intelligence in pancreatic cancer. Front Oncol. 2024;14:1345810. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 11] [Reference Citation Analysis (0)] |
| 237. | Placido D, Yuan B, Hjaltelin JX, Zheng C, Haue AD, Chmura PJ, Yuan C, Kim J, Umeton R, Antell G, Chowdhury A, Franz A, Brais L, Andrews E, Marks DS, Regev A, Ayandeh S, Brophy MT, Do NV, Kraft P, Wolpin BM, Rosenthal MH, Fillmore NR, Brunak S, Sander C. A deep learning algorithm to predict risk of pancreatic cancer from disease trajectories. Nat Med. 2023;29:1113-1122. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 92] [Cited by in RCA: 143] [Article Influence: 71.5] [Reference Citation Analysis (0)] |
| 238. | Liao J, Li X, Gan Y, Han S, Rong P, Wang W, Li W, Zhou L. Artificial intelligence assists precision medicine in cancer treatment. Front Oncol. 2022;12:998222. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 46] [Cited by in RCA: 79] [Article Influence: 39.5] [Reference Citation Analysis (0)] |
| 239. | Shahzad K, Abu-Zanona M, Elzaghmouri BM, AbdelRahman SM, Fadol Osman AA, Al-Khateeb A, Khatoon F. USING ARTIFICIAL INTELLIGENCE AND MACHINE LEARNING APPROACHES TO ENHANCE CANCER THERAPY AND DRUG DISCOVERY: A NARRATIVE REVIEW. J Ayub Med Coll Abbottabad. 2024;36:183-189. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
| 240. | Cai Z, Poulos RC, Liu J, Zhong Q. Machine learning for multi-omics data integration in cancer. iScience. 2022;25:103798. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 102] [Cited by in RCA: 139] [Article Influence: 46.3] [Reference Citation Analysis (0)] |
| 241. | Katta MR, Kalluru PKR, Bavishi DA, Hameed M, Valisekka SS. Artificial intelligence in pancreatic cancer: diagnosis, limitations, and the future prospects-a narrative review. J Cancer Res Clin Oncol. 2023;149:6743-6751. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 10] [Cited by in RCA: 9] [Article Influence: 4.5] [Reference Citation Analysis (0)] |
| 242. | Correction: Quality standards in upper gastrointestinal endoscopy: a position statement of the British Society of Gastroenterology (BSG) and Association of Upper Gastrointestinal Surgeons of Great Britain and Ireland (AUGIS). Gut. 2017;66:2188. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Cited by in RCA: 1] [Article Influence: 0.1] [Reference Citation Analysis (0)] |
| 243. | Correction to Lancet Gastroenterol Hepatol 2020; 5: 352-61. Lancet Gastroenterol Hepatol. 2020;5:e3. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
| 244. | Rahib L, Coffin T, Kenner B. Factors Driving Pancreatic Cancer Survival Rates. Pancreas. 2025;54:e530-e536. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 4] [Reference Citation Analysis (0)] |
| 245. | Hartupee C, Nagalo BM, Chabu CY, Tesfay MZ, Coleman-Barnett J, West JT, Moaven O. Pancreatic cancer tumor microenvironment is a major therapeutic barrier and target. Front Immunol. 2024;15:1287459. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2] [Cited by in RCA: 44] [Article Influence: 44.0] [Reference Citation Analysis (0)] |
