1
|
Li F, Shen F. Metastatic pancreatic cancer with activating BRAF V600E mutations: A case report. World J Clin Cases 2025; 13:101665. [DOI: 10.12998/wjcc.v13.i16.101665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/06/2024] [Accepted: 01/11/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is a highly malignant tumor that is resistant to chemotherapy, radiotherapy and immunotherapy. Combination chemotherapy regimens are the standard first-line regimens for metastatic disease, with a median survival < 12 months. Although recurrent genomic alterations such as the BRAF V600E mutation have been reported in PC, evidence supporting the clinical effectiveness of molecularly guided targeted therapies is limited.
CASE SUMMARY We report a case of a 33-year-old male who was referred to our department with weight loss of 5 kg in 2 months, anorexia and abdominal pain. Imaging showed extensive lesions involving the pancreas, liver, bones, muscles and lymph nodes accompanied by elevated carbohydrate antigen 19-9 (CA19-9) and carcinoembryonic antigen (CEA). Biopsy yielded a diagnosis of PC. Treatment with gemcitabine and nab-paclitaxel was initiated, but the disease progressed in < 2 months even though the patient’s general condition improved. Molecular testing revealed the presence of BRAF mutation. Dabrafenib/trametinib combination therapy was introduced, and the patient was treated for 2 months with a decrease in CA19-9 and CEA levels, but he died after 2 months of treatment.
CONCLUSION BRAF alterations are infrequent in PC. This case highlights the significance of molecular profiling in patients with PC, especially in patients with a high tumor burden.
Collapse
Affiliation(s)
- Fang Li
- Department of Medical Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, Fujian Province, China
- Xiamen Clinical Research Center for Cancer, Xiamen 361015, Fujian Province, China
- Clinical Research Center for Precision Medicine of Abdominal Tumor of Fujian Province, Xiamen 361015, Fujian Province, China
| | - Feng Shen
- Department of Medical Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, Fujian Province, China
- Xiamen Clinical Research Center for Cancer, Xiamen 361015, Fujian Province, China
- Clinical Research Center for Precision Medicine of Abdominal Tumor of Fujian Province, Xiamen 361015, Fujian Province, China
| |
Collapse
|
2
|
Matejcic M, Teer JK, Hoehn HJ, Diaz DB, Shankar K, Gong J, Nguyen NT, Loroña NC, Coppola D, Fulmer CG, Saglam O, Jiang K, Cress WD, Muñoz-Antonia T, Flores I, Gordián ER, Oliveras Torres JA, Felder SI, Sanchez J, Fleming JB, Siegel EM, Freedman JA, Dutil J, Stern MC, Fridley BL, Figueiredo JC, Schmit SL. Colorectal Tumors in Diverse Patient Populations Feature a Spectrum of Somatic Mutational Profiles. Cancer Res 2025; 85:1928-1944. [PMID: 40126181 DOI: 10.1158/0008-5472.can-24-0747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/21/2024] [Accepted: 02/25/2025] [Indexed: 03/25/2025]
Abstract
Admixed populations, including the Hispanic/Latino/a community, are underrepresented in cancer genetic/genomic studies. Leveraging the Latino Colorectal Cancer Consortium (LC3) and other existing datasets, we analyzed whole-exome sequencing data on tumor/normal pairs from 718 individuals with colorectal cancer to map somatic mutational features by ethnicity and genetic similarity. Global proportions of African, Asian, European, and Native American genetic ancestries were estimated using ADMIXTURE. Associations between these proportions and somatic mutational features were examined using logistic regression. APC, TP53, and KRAS were the top three mutated genes across all participants and in the subset of Latino individuals in LC3. In analyses examining recurrently mutated genes, tumors from patients of Latino ethnicity had fewer KRAS and PIK3CA mutations compared with tumors from non-Latino patients. Genetic ancestry overall was associated with CDC27 mutation status, and African genetic ancestry was associated with SMAD2 mutation status. In exome-wide analyses, African genetic ancestry was significantly associated with higher odds of mutation in KNCN and TMEM184B. Native American genetic ancestry was associated with a lower frequency of microsatellite instability-high tumors. The SBS11 mutational signature was associated with Native American genetic ancestry as well as Latino ethnicity. In an independent replication dataset, MSK-IMPACT, estimates of association were largely consistent in direction but nonsignificant. A meta-analysis of LC3 and MSK-IMPACT showed that African genetic ancestry was significantly associated with KRAS mutation status and MSI status. This work facilitates precision medicine initiatives by providing insights into the contribution of genetic ancestry to molecular features of colorectal tumors. Significance: Analysis of tumors from various populations can broadly characterize genomic landscapes and enhance precision medicine strategies.
Collapse
Affiliation(s)
- Marco Matejcic
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jamie K Teer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Hannah J Hoehn
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Non-Therapeutic Research Office, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Diana B Diaz
- Non-Therapeutic Research Office, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Kritika Shankar
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jun Gong
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Nathalie T Nguyen
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Nicole C Loroña
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Domenico Coppola
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Clifton G Fulmer
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Ozlen Saglam
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Kun Jiang
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - W Douglas Cress
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Teresita Muñoz-Antonia
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Idhaliz Flores
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Edna R Gordián
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - José A Oliveras Torres
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Seth I Felder
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Julian Sanchez
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jason B Fleming
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Erin M Siegel
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Non-Therapeutic Research Office, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jennifer A Freedman
- Division of Medical Oncology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
- Duke Cancer Institute, Durham, North Carolina
| | - Julie Dutil
- Division of Clinical and Translational Cancer Research, Comprehensive Cancer Center of the University of Puerto Rico, San Juan, Puerto Rico
| | - Mariana C Stern
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, California
| | - Brooke L Fridley
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jane C Figueiredo
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Stephanie L Schmit
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
3
|
Preckwinkel P, Mir KUI, Otto FW, Elrewany H, Sinz A, Hüttelmaier S, Bley N, Gutschner T. Long Non-Coding RNAs and RNA-Binding Proteins in Pancreatic Cancer Development and Progression. Cancers (Basel) 2025; 17:1601. [PMID: 40427100 PMCID: PMC12110025 DOI: 10.3390/cancers17101601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer and is responsible for about 467,000 cancer deaths annually. An oftentimes asymptomatic early phase of this disease results in a delayed diagnosis, and patients often present with advanced disease. Current treatment options have limited survival benefits, and only a minor patient population carries actionable genomic alterations. Hence, innovative personalized treatment strategies that consider molecular, cellular and functional analyses are urgently needed for pancreatic cancer patients. However, the majority of the genetic alterations found in PDAC are currently undruggable, or patients' response is not as expected. Therefore, non-genomic biomarkers and alternative molecular targets should be considered in order to advance the clinical management of PDAC patients. In line with this, recent gene expression and single-cell transcriptome analyses have identified molecular subtypes and transcriptional cell states that affect disease progression and drug efficiency. In this review, we will introduce long non-coding RNAs (lncRNAs) as well as RNA-binding proteins (RBPs) that are able to modulate the transcriptome of a cell through diverse mechanisms, thereby contributing to disease progression. We will provide a brief overview about the general functions of lncRNAs and RBPs, respectively. Subsequently, we will highlight selected lncRNAs and RBPs that have been shown to play a role in PDAC development, progression and drug response. Finally, we will present strategies aiming to interfere with the expression and function of lncRNAs and RBPs.
Collapse
Affiliation(s)
- Pit Preckwinkel
- Section for RNA Biology and Pathogenesis, Institute of Molecular Medicine, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Khursheed Ul Islam Mir
- Section for Molecular Cell Biology, Institute of Molecular Medicine, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (K.U.I.M.); (H.E.); (S.H.)
| | - Florian W. Otto
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (F.W.O.); (A.S.)
- Center for Structural Mass Spectrometry, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Hend Elrewany
- Section for Molecular Cell Biology, Institute of Molecular Medicine, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (K.U.I.M.); (H.E.); (S.H.)
| | - Andrea Sinz
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (F.W.O.); (A.S.)
- Center for Structural Mass Spectrometry, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Stefan Hüttelmaier
- Section for Molecular Cell Biology, Institute of Molecular Medicine, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (K.U.I.M.); (H.E.); (S.H.)
| | - Nadine Bley
- Section for Molecular Cell Biology, Institute of Molecular Medicine, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (K.U.I.M.); (H.E.); (S.H.)
| | - Tony Gutschner
- Section for RNA Biology and Pathogenesis, Institute of Molecular Medicine, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| |
Collapse
|
4
|
Awosika JA, Gulley JL, Pastor DM. Deficient Mismatch Repair and Microsatellite Instability in Solid Tumors. Int J Mol Sci 2025; 26:4394. [PMID: 40362635 PMCID: PMC12072705 DOI: 10.3390/ijms26094394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/26/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
The integrity of the genome is maintained by mismatch repair (MMR) proteins that recognize and repair base mismatches and insertion/deletion errors generated during DNA replication and recombination. A defective MMR system results in genome-wide instability and the progressive accumulation of mutations. Tumors exhibiting deficient MMR (dMMR) and/or high levels of microsatellite instability (termed "microsatellite instability high", or MSI-H) have been shown to possess fundamental differences in clinical, pathological, and molecular characteristics, distinguishing them from their "microsatellite stable" (MSS) counterparts. Molecularly, they are defined by a high mutational burden, genetic instability, and a distinctive immune profile. Their distinct genetic and immunological profiles have made dMMR/MSI-H tumors particularly amenable to treatment with immune checkpoint inhibitors (ICIs). The ongoing development of biomarker-driven therapies and the evaluation of novel combinations of immune-based therapies, with or without the use of conventional cytotoxic treatment regimens, continue to refine treatment strategies with the goals of maximizing therapeutic efficacy and survival outcomes in this distinct patient population. Moreover, the resultant knowledge of the mechanisms by which these features are suspected to render these tumors more responsive, overall, to immunotherapy may provide information regarding the potential optimization of this therapeutic approach in tumors with proficient MMR (pMMR)/MSS tumors.
Collapse
Affiliation(s)
- Joy A. Awosika
- Gastrointestinal Malignancies Section, Thoracic & GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - James L. Gulley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Danielle M. Pastor
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
5
|
Eikenboom EL, Nasar N, Seier K, Gönen M, Spaander MCW, O'Reilly EM, Jarnagin WR, Drebin J, D'Angelica MI, Kingham TP, Balachandran VP, Soares KC, Wagner A, Wei AC. Survival of Patients with Resected Microsatellite Instability-High, Mismatch Repair Deficient, and Lynch Syndrome-Associated Pancreatic Ductal Adenocarcinomas. Ann Surg Oncol 2025; 32:3568-3577. [PMID: 39656390 DOI: 10.1245/s10434-024-16621-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 11/19/2024] [Indexed: 04/24/2025]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains a challenging disease due to its aggressiveness, late-stage diagnosis, and limited treatment options. Microsatellite instability-high (MSI-H) cancers are susceptible to immune checkpoint inhibitors. Survival outcomes for patients with MSI-H PDAC are unknown as the disease is rare. METHODS This study included patients with PDACs surgically resected from 1990 to 2023, and those with germline or sporadic pathogenic variants in DNA mismatch repair genes were identified. The study matched MSI-H, mismatch repair-deficient (MMRd), and Lynch syndrome (LS)-associated PDAC cases (on age, gender, and year of surgery) with microsatellite-stable (MSS), mismatch repair-proficient, or non-LS-associated PDAC cases in a 1:2 ratio. A generalized estimating equation Cox model with a robust sandwich estimator was used to compare overall survival (OS) in the matched cohorts. RESULTS Of 936 cases, 18 were included. Eight cases were MSI-H/MMRd, two were MSI/IHC-indeterminate, seven were MSS, and one was not tested for MSI. Nine patients had LS (MLH1 [n = 1], MSH2 [n = 4], MSH6 [n = 1], PMS2 [n = 3]), and nine patients had sporadic pathogenic variants in DNA MMR genes (MLH1 [n = 4], MSH6 [n = 5]). After matching to 36 control patients, the MSI-H/MMRd/LS PDACs had a significantly better OS (hazard ratio [HR], 0.36 [95% confidence interval [CI], 0.18-0.73; p = 0.005]; 5-year OS: MSI-H 77% [95% CI 58-100%] vs. MSS 27% [95% CI 15-51%]). CONCLUSION Before routine use of immune checkpoint inhibitors, the patients with MSI-H, MMRd, and LS-associated PDACs displayed significantly better survival than the patients with MSS, MMR-proficient, non-LS-associated PDACs. It is expected that survival for this cohort will further improve with increased availability of immunotherapy.
Collapse
Affiliation(s)
- Ellis L Eikenboom
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Clinical Genetics, Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Naaz Nasar
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenneth Seier
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Manon C W Spaander
- Department of Gastroenterology and Hepatology, Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William R Jarnagin
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeffrey Drebin
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael I D'Angelica
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - T Peter Kingham
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vinod P Balachandran
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin C Soares
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anja Wagner
- Department of Clinical Genetics, Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Alice C Wei
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
6
|
Chung V, Mizrahi JD, Pant S. Novel Therapies for Pancreatic Cancer. JCO Oncol Pract 2025; 21:613-619. [PMID: 39591547 DOI: 10.1200/op.24.00279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 10/15/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Pancreatic adenocarcinoma (PDAC) unfortunately remains a highly fatal disease with a 5-year survival rate of only 11%. If surgical resection is not possible, systemic chemotherapy represents the standard-of-care approach to management. Combination chemotherapy regimens using fluorouracil (fluorouracil, oxaliplatin, leucovorin, and irinotecan; and fluorouracil, leucovorin, and oxaliplatin) or gemcitabine with albumin-bound paclitaxel have the potential to improve overall survival for patients with advanced disease. With the increasing understanding of the molecular drivers of pancreatic cancer, novel therapeutic approaches have made incremental progress for these patients. The molecular landscape of PDAC has been studied extensively. Approximately 90% of PDACs harbor mutations in the KRAS gene, a driver mutation that has long been considered undruggable. However, novel KRAS inhibitors have shown therapeutic promise in early-phase clinical trials, with larger studies ongoing. Less frequently encountered genomic aberrations that have therapeutic potential include NRG, BRAF, NTRK, HER2, BRCA, PALB2, and claudin. Immune checkpoint inhibitors have unfortunately yielded disappointing efficacy for the majority of patients with pancreatic cancer, except for those with tumors exhibiting deficiency in mismatch repair proteins. Alternative approaches to incorporate immunotherapy have shown more promise such as use of immune checkpoint inhibitors for selected patients in the maintenance setting and potential vaccine therapies in the postsurgery adjuvant setting. It is vital to perform molecular profiling in all patients with PDAC to identify potential treatment targets, and to enroll patients in clinical trials whenever possible.
Collapse
Affiliation(s)
- Victoria Chung
- Ochsner Medical Center, New Orleans, LA
- MD Anderson Cancer Center, Houston, TX
| | | | | |
Collapse
|
7
|
Doi T, Ishikawa T, Moriguchi M, Itoh Y. Current status of cancer genome medicine for pancreatic ductal adenocarcinoma. Jpn J Clin Oncol 2025; 55:443-452. [PMID: 39893577 DOI: 10.1093/jjco/hyaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis; however, advancements in cancer genome profiling using next-generation sequencing have provided new perspectives. KRAS mutations are the most frequently observed genomic alterations in patients with PDAC. However, until recently, it was not considered a viable therapeutic target. Although KRAS G12C mutations for which targeted therapies are already available are infrequent in PDAC, treatments targeting KRAS G12D and pan-KRAS are still under development. Similarly, new treatment methods for KRAS, such as chimeric antigen receptor T-cell therapy, have been developed. Several other potential therapeutic targets have been identified for KRAS wild-type PDAC. For instance, immune checkpoint inhibitors have demonstrated efficacy in PDAC treatment with microsatellite instability-high/deficient mismatch repair and tumor mutation burden-high profiles. However, for other PDAC cases with low immunogenicity, combination therapies that enhance the effectiveness of immune checkpoint inhibitors are being considered. Additionally, homologous recombination repair deficiencies, including BRCA1/2 mutations, are prevalent in PDAC and serve as important biomarkers for therapies involving poly (adenosine diphosphate-ribose) polymerase inhibitors and platinum-based therapies. Currently, olaparib is available for maintenance therapy of BRCA1/2 mutation-positive PDAC. Further therapeutic developments are ongoing for genetic abnormalities involving BRAF V600E and the fusion genes RET, NTRK, NRG, ALK, FGFR2, and ROS1. Overcoming advanced PDAC remains a formidable challenge; however, this review outlines the latest therapeutic strategies that are expected to lead to significant advancements.
Collapse
Affiliation(s)
- Toshifumi Doi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Medical Oncology Unit, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Michihisa Moriguchi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
8
|
Liang KL, Azad NS. Immune-Based Strategies for Pancreatic Cancer in the Adjuvant Setting. Cancers (Basel) 2025; 17:1246. [PMID: 40227779 PMCID: PMC11988091 DOI: 10.3390/cancers17071246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related mortality in the United States, with poor overall survival across all stages. Less than 20% of patients are eligible for curative surgical resection at diagnosis, and despite adjuvant chemotherapy, most will experience disease recurrence within two years. The incorporation of immune-based strategies in the adjuvant setting remains an area of intense investigation with unrealized promise. It offers the potential of providing durable disease control for micro-metastatic disease following curative intent surgery and enabling personalized treatments based on mutational neoantigen profiles derived from resected specimens. However, most of these attempts have failed to demonstrate significant clinical success, likely due to the immunosuppressive tumor microenvironment (TME) and individual genetic heterogeneity. Despite these challenges, immune-based strategies, such as therapeutic vaccines targeted towards neoantigens, have demonstrated promise via immune activation and induction of T-cell tumor infiltration. In this review, we will highlight the foundational lessons learned from previous clinical trials of adjuvant immunotherapy, discussing the knowledge gained from analyses of trials with disappointing results. In addition, we will discuss how these data have been incorporated to design new agents and study concepts that are proving to be exciting in more recent trials, such as shared antigen vaccines and combination therapy with immune-checkpoint inhibitors and chemotherapy. This review will evaluate novel approaches in ongoing and future clinical studies and provide insight into how these immune-based strategies might evolve to address the unique challenges for treatment of PDAC in the adjuvant setting.
Collapse
Affiliation(s)
| | - Nilofer S. Azad
- Department of Oncology, Sidney Kimmel Comprehensive Cancer, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| |
Collapse
|
9
|
So E, Hayashi H, Shimozaki K, Horie S, Kishimoto S, Chida A, Saito Y, Tsugaru K, Hirata K, Tanishima S, Nishihara H, Kanai T, Hamamoto Y. Clinical utility of comprehensive genomic profiling for advanced pancreatic cancer: insights from real-world data analysis. Int J Clin Oncol 2025; 30:728-737. [PMID: 39961905 DOI: 10.1007/s10147-025-02713-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 01/23/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Precision medicine is a promising therapeutic strategy for pancreatic cancer. However, only a few patients are eligible for genotype-matched treatments because of the low detection rate of actionable genomic alterations, and the clinical application of comprehensive genomic profiling (CGP) in pancreatic cancer has not been completely investigated. CGP provides considerable information, including the prognosis and eligibility of patients for genotype-matched treatments, which can guide physicians' treatment strategies. This study aimed to investigate the contribution of CGP to patient outcomes. METHODS This single-center retrospective cohort study enrolled patients with recurrent or metastatic pancreatic cancer with adenocarcinoma or adenosquamous carcinoma who underwent systemic chemotherapy between April 2018 and April 2022. We reviewed the medical records for patient characteristics, survival, and genomic information. We compared overall survival (OS) between patients who received CGP (CGP group) and those who did not (non-CGP group). RESULTS Of 111 eligible patients, 59 underwent CGP. Median OS was significantly longer in the CGP than the non-CGP group (25.2 vs. 11.8 months; hazard ratio, 0.49; P = 0.0013). Six patients (10.2%) underwent genotype-matched treatments, with a median OS of 35.5 months, compared to 17.0 months for those who did not. The CGP group demonstrated a higher transition rate to subsequent chemotherapy than did the non-CGP group (76.3% vs. 48.1%, P = 0.0030). CONCLUSIONS OS was prolonged in patients with pancreatic cancer who underwent CGP, probably due to its influence on physicians' treatment strategies. These findings highlight the importance of the proactive and timely implementation of CGP in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Eiichiro So
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hideyuki Hayashi
- Keio Cancer Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Keitaro Shimozaki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Sara Horie
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shotaro Kishimoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Akihiko Chida
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yuki Saito
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kai Tsugaru
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kenro Hirata
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shigeki Tanishima
- Department of Biomedical Informatics Development, Mitsubishi Electric Software Co., Ltd, Tokyo, Japan
| | - Hiroshi Nishihara
- Keio Cancer Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yasuo Hamamoto
- Keio Cancer Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
10
|
Ohtsubo K, Sato S, Sakaguchi H, Kotani H, Nishiyama A, Yamashita K, Yano S, Toshima F, Inoue D, Gabata T, Ikeda H, Watanabe A, Notohara K, Fujisawa T, Nakamura Y, Yoshino T, Miyake K, Miwa K, Takeuchi S. Case Report: Medullary carcinoma of the pancreas with MLH1 promoter hypermethylation, induced deficient mismatch repair, successfully treated with an immune checkpoint inhibitor. Front Oncol 2025; 15:1551038. [PMID: 40236650 PMCID: PMC11997870 DOI: 10.3389/fonc.2025.1551038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
We report the case of a 75-year-old woman with a pancreatic body mass. Pathological findings from endoscopic ultrasonography-guided fine-needle aspiration revealed medullary carcinoma of the pancreas (MCP). Deficient mismatch repair (dMMR) and high microsatellite instability (MSI-H) were identified through immunohistochemistry and next generation sequencing, respectively. While immunohistochemistry suggested MLH1 abnormality, no MLH1 mutation was; hypermethylation of the MLH1 promoter was later confirmed via bisulfite sequencing. The patient initially received nab-paclitaxel plus gemcitabine, achieving tumor shrinkage. Upon tumor regrowth, she was treated with the anti-programmed cell death-1 immune checkpoint inhibitor (ICI) pembrolizumab, which resulted in significant tumor reduction. This is the first case report of MCP with dMMR/MSI-H due to MLH1 promoter hypermethylation, effectively treated with an ICI.
Collapse
Affiliation(s)
- Koushiro Ohtsubo
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Shigeki Sato
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Hiroyuki Sakaguchi
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Hiroshi Kotani
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Akihiro Nishiyama
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Kaname Yamashita
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Seiji Yano
- Department of Respiratory Medicine, Kanazawa University Hospital, Kanazawa, Japan
| | - Fumihito Toshima
- Department of Radiology, Kanazawa University Hospital, Kanazawa, Japan
| | - Dai Inoue
- Department of Radiology, Kanazawa University Hospital, Kanazawa, Japan
| | - Toshifumi Gabata
- Department of Radiology, Kanazawa University Hospital, Kanazawa, Japan
| | - Hiroko Ikeda
- Division of Human Pathology, Kanazawa University Hospital, Kanazawa, Japan
| | - Atsushi Watanabe
- Division of Clinical Genetics, Kanazawa University Hospital, Kanazawa, Japan
| | - Kenji Notohara
- Department of Anatomic Pathology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Takao Fujisawa
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- International Research Promotion Office, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshiaki Nakamura
- International Research Promotion Office, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- International Research Promotion Office, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kunio Miyake
- Department of Epidemiology and Environmental Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Japan
| | - Kazuhiro Miwa
- Internal Medicine, Komatsu Municipal Hospital, Komatsu, Japan
| | - Shinji Takeuchi
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| |
Collapse
|
11
|
Matsubayashi H, Kiyozumi Y, Ono H. Genetic medicine of familial and hereditary pancreatic cancer: Recent update in the era of precision cancer medicine. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2025; 32:212-227. [PMID: 39814596 DOI: 10.1002/jhbp.12112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
In Japan, 5 years have passed since the initiation of precision cancer medicine, and recent data accumulation in familial pancreatic cancer (FPC) and hereditary pancreatic cancer is outstanding. Multigene germline panel tests (MGPTs) have revealed that 7%-18% of patients with pancreatic cancer (PC) harbor pathogenic germline variants (PGVs), almost equal to the levels of breast, ovarian, endometrial, and colorectal cancers, with a higher incidence in FPC (14%-26%). The majority of PGVs seen in PC patients are clinically actionable and associated with homologous recombination (HR) pathways (6%-10%, particularly BRCA1/2 in 5%-6%), and the clinical guidelines recommend or propose genetic testing for all PC patients. Consensus guidelines have been established for most of the hereditary syndromes associated with PC risks, and surveillances of the pancreas and other at-risk organs are recommended for PGV carriers. Hereditary breast and ovarian cancer (HBOC) is the commonest hereditary cancer syndrome that has moderately increasing life-time risks of PC (3%-7% in Western countries); however, recent Japanese research demonstrated a higher risk level (BRCA1: 16%, BRCA2: 14%). Moreover, recent evidence has suggested a risk linkage between PC and ovarian cancer in HBOC pedigrees. High scores of homologous recombination deficiency suggest biallelic dysfunction of BRCA or other HR-related genes, and the likely effectiveness of platinum agents and PARP inhibitors against PCs. Remote counseling and testing are possible option in the future genetic medicine. As PC ranks in the second commonest target of precision cancer medicine in Japan, we must treat the patients and manage their at-risk relatives efficiently.
Collapse
Affiliation(s)
- Hiroyuki Matsubayashi
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, Shizuoka, Japan
- Division of Endoscopy, Shizuoka Cancer Center, Shizuoka, Japan
| | - Yoshimi Kiyozumi
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, Shizuoka, Japan
| | - Hiroyuki Ono
- Division of Endoscopy, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
12
|
Kalfakakou D, Cameron DC, Kawaler EA, Tsuda M, Wang L, Jing X, Hajdu C, Tamayo DL, Shim Y, Ackermann A, Weissinger D, Zimny H, Hernandez R, Beier M, Dimartino D, Meyn P, Rice K, Selvaraj S, Loomis C, Heguy A, Lund AW, Sears RC, Welling TH, Dolgalev I, Tsirigos A, Simeone DM. Clonal Heterogeneity in Human Pancreatic Ductal Adenocarcinoma and Its Impact on Tumor Progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637729. [PMID: 39990481 PMCID: PMC11844494 DOI: 10.1101/2025.02.11.637729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer characterized by profound desmoplasia and cellular heterogeneity, which cannot be fully resolved using traditional bulk sequencing approaches. To understand the contribution of this heterogeneity to PDAC biology, we analyzed a large cohort of primary human PDAC samples (n = 62), profiling 443,451 single cells and 53,236 spatial transcriptomic spots using a combined single-cell RNA sequencing and spatial transcriptomics approach. Our analysis revealed significant intratumoral heterogeneity, with multiple genetically distinct neoplastic clones co-existing within individual tumors. These clones exhibited diverse transcriptional states and subtype profiles, challenging the traditional binary classification of PDAC into basal and classical subtypes; instead, our findings support a transcriptional continuum influenced by clonal evolution and spatial organization. Additionally, these clones each interacted uniquely with surrounding cell types in the tumor microenvironment. Phylogenetic analysis uncovered a rare but consistent classical-to-basal clonal transition associated with MYC amplification and immune response depletion, which were validated experimentally, suggesting a mechanism driving the emergence of a more aggressive basal clonal phenotype. Spatial analyses further revealed dispersed clones enriched for epithelial-to-mesenchymal transition (EMT) activity and immune suppression, correlating with metastatic potential and colonization of lymph node niches. These dispersed clones tended to transition toward a basal phenotype, contributing to disease progression. Our findings highlight the critical role of clonal diversity, transcriptional plasticity, and TME interactions in shaping human PDAC biology. This work provides new insights into the molecular and spatial heterogeneity of PDAC and offers potential avenues for therapeutic intervention targeting clonal evolution and the mechanisms driving metastasis.
Collapse
|
13
|
Varghese AM, Perry MA, Chou JF, Nandakumar S, Muldoon D, Erakky A, Zucker A, Fong C, Mehine M, Nguyen B, Basturk O, Balogun F, Kelsen DP, Brannon AR, Mandelker D, Vakiani E, Park W, Yu KH, Stadler ZK, Schattner MA, Jarnagin WR, Wei AC, Chakravarty D, Capanu M, Schultz N, Berger MF, Iacobuzio-Donahue CA, Bandlamudi C, O'Reilly EM. Clinicogenomic landscape of pancreatic adenocarcinoma identifies KRAS mutant dosage as prognostic of overall survival. Nat Med 2025; 31:466-477. [PMID: 39753968 PMCID: PMC11835752 DOI: 10.1038/s41591-024-03362-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/17/2024] [Indexed: 01/11/2025]
Abstract
Nearly all pancreatic adenocarcinomas (PDAC) are genomically characterized by KRAS exon 2 mutations. Most patients with PDAC present with advanced disease and are treated with cytotoxic therapy. Genomic biomarkers prognostic of disease outcomes have been challenging to identify. Herein leveraging a cohort of 2,336 patients spanning all disease stages, we characterize the genomic and clinical correlates of outcomes in PDAC. We show that a genomic subtype of KRAS wild-type tumors is associated with early disease onset, distinct somatic and germline features, and significantly better overall survival. Allelic imbalances at the KRAS locus are widespread. KRAS mutant allele dosage gains, observed in one in five (20%) KRAS-mutated diploid tumors, are correlated with advanced disease and demonstrate prognostic potential across disease stages. With the rapidly expanding landscape of KRAS targeting, our findings have potential implications for clinical practice and for understanding de novo and acquired resistance to RAS therapeutics.
Collapse
Affiliation(s)
- Anna M Varghese
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Maria A Perry
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Joanne F Chou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Subhiksha Nandakumar
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Daniel Muldoon
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Amanda Erakky
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Amanda Zucker
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Christopher Fong
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Miika Mehine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Bastien Nguyen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Olca Basturk
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Fiyinfolu Balogun
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - David P Kelsen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - A Rose Brannon
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Diana Mandelker
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Efsevia Vakiani
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Wungki Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Kenneth H Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Mark A Schattner
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - William R Jarnagin
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Alice C Wei
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Debyani Chakravarty
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Marinela Capanu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Nikolaus Schultz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Michael F Berger
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Christine A Iacobuzio-Donahue
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Chaitanya Bandlamudi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| |
Collapse
|
14
|
Robinson RM, Reyes L, Christopher BN, Duncan RM, Burge RA, Siegel J, Nasarre P, Wang P, O'Bryan JP, Hobbs GA, Klauber-DeMore N, Dolloff NG. A High-Affinity Monoclonal Antibody Against the Pancreatic Ductal Adenocarcinoma Target, Anterior Gradient-2 (AGR2/PDIA17). Antibodies (Basel) 2024; 13:101. [PMID: 39727484 DOI: 10.3390/antib13040101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Anterior Gradient-2 (AGR2/PDIA17) is a member of the protein disulfide isomerase (PDI) family of oxidoreductases. AGR2 is up-regulated in several solid tumors, including pancreatic ductal adenocarcinoma (PDAC). Given the dire need for new therapeutic options for PDAC patients, we investigated the expression and function of AGR2 in PDAC and developed a novel series of affinity-matured AGR2-specific single-chain variable fragments (scFvs) and monoclonal antibodies. RESULTS We found that AGR2 was expressed in approximately 90% of PDAC but not normal pancreas biopsies, and the level of AGR2 expression correlated with increasing disease stage. AGR2 expression was inversely related to SMAD4 status in PDAC and colorectal cancer cell models and was secreted from cells into their media. In normal tissues, a high density of AGR2 was detected in the epithelium of cells in the digestive tract but was lacking in most other normal tissue systems. The addition of recombinant AGR2 to cell culture and genetic overexpression of AGR2 increased the adhesion, motility, and invasiveness of both human and mouse PDAC cells. Human phage display library screening led to the discovery of multiple AGR2-specific scFv clones that were affinity-matured to produce monoclonal antibody (MAb) clones with low picomolar binding affinity (S31R/A53F/Y). These high-affinity MAbs inhibited AGR2-mediated cell adhesion, migration, and binding to LYPD3, which is a putative cell surface binding partner of AGR2. CONCLUSIONS Our study provides novel, high-affinity, fully human, anti-AGR2 MAbs that neutralize the pro-tumor effects of extracellular AGR2 in PDAC.
Collapse
Affiliation(s)
- Reeder M Robinson
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Leticia Reyes
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Benjamin N Christopher
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ravyn M Duncan
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Rachel A Burge
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Julie Siegel
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Patrick Nasarre
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | - John P O'Bryan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - G Aaron Hobbs
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Nancy Klauber-DeMore
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
- MUSC Hollings Cancer Center, Charleston, SC 29425, USA
| | - Nathan G Dolloff
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
- MUSC Hollings Cancer Center, Charleston, SC 29425, USA
- Zucker Institute for Innovation Commercialization, Charleston, SC 29425, USA
| |
Collapse
|
15
|
Li KY, Lowy AM, Fanta P. Quantification of PD-L1 expression and tumor mutational burden in biologically distinct advanced pancreatic cancers responding to pembrolizumab: case reports. Front Immunol 2024; 15:1452543. [PMID: 39687619 PMCID: PMC11646977 DOI: 10.3389/fimmu.2024.1452543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
Background The advent of checkpoint therapy is one of the most important recent advancements in cancer therapy. Though checkpoint therapy is a mainstay in some cancers, it has been largely ineffective in treating cancers of the pancreas. Pancreatic ductal adenocarcinoma and pancreatic neuroendocrine tumors are seldom responsive to checkpoint inhibition. Case presentations Here we present two cases of advanced pancreatic cancers that either failed to respond or recurred following conventional treatments. Tissue from each tumor was sequenced and analyzed for PD-L1 expression. Each patient was started on checkpoint blockade after assessing for a predictive biomarker, either the combined positive score or the tumor mutational burden. In each case, checkpoint blockade led to durable radiographic responses. Conclusions We therefore propose that it is reasonable to assess combined positive score and tumor mutational burden in refractory or recurrent pancreatic cancers when initiation of ICB is being considered.
Collapse
Affiliation(s)
- Kevin Y. Li
- Department of Surgery, Division of Surgical Oncology, University of California, San
Diego, San Diego, CA, United States
- Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Andrew M. Lowy
- Department of Surgery, Division of Surgical Oncology, University of California, San
Diego, San Diego, CA, United States
- Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Paul Fanta
- Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
- Division of Hematology Oncology, Department of Medicine, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
16
|
O’Connor CA, Harrold E, Lin D, Walch H, Gazzo A, Ranganathan M, Kane S, Keane F, Schoenfeld J, Moss D, Thurtle-Schmidt DM, Suehnholz SP, Chakravarty D, Balogun F, Varghese A, Yu K, Kelsen D, Latham A, Weigelt B, Park W, Stadler Z, O’Reilly EM. Lynch Syndrome and Somatic Mismatch Repair Variants in Pancreas Cancer. JAMA Oncol 2024; 10:1511-1518. [PMID: 39235819 PMCID: PMC11378065 DOI: 10.1001/jamaoncol.2024.3651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 05/20/2024] [Indexed: 09/06/2024]
Abstract
Importance Microsatellite (MS) instability (MSI-H) occurs frequently in Lynch syndrome (LS)-associated tumors and is associated with response to immune checkpoint blockade (ICB) therapy. MSI-H is conferred by germline or somatic variants in mismatch repair genes. The contribution of somatic oncogenesis to MSI-H in pancreatic cancer (PC) is unknown. Objective To evaluate an LS-related PC cohort to define clinicogenomic features, describe somatic MSI-H cases (germline negative), characterize response to ICB, and guide preferred MS testing methods. Design, Setting, and Participants This single-institution, retrospective analysis was conducted from March 2012 to July 2023 at Memorial Sloan Kettering Cancer Center and included 55 patients with PC and either an LS germline pathogenic variant (gPV) or somatic mismatch repair (MMR) variant. Main Outcomes and Measures Composite MMR and MS status determined using orthogonal methods. An artificial intelligence classifier was used to account for low-cellularity specimens. Demographic and clinical data were abstracted from medical record. Zygosity status and somatic comutation landscape analyzed. Results Fifty-five patients (23 women [42%]) had PC and an MMR variant: 32 (58%) had LS (LS cohort) and 23 (42%) had a somatic MMR variant (no germline pathogenic variant, somatic MMR cohort). In the LS cohort, 10 (31%) had gMSH2, 9 (28%) gMSH6, 8 (25%) gPMS2, 4 (13%) gMLH1, 1 (3%) gEPCAM. The median age at diagnosis was 68 years (range, 45-88 years). For composite MS status, 17 (59%) were MSI-H, 12 (41%) MS stable, and 3 MS unknown. Five cases were reclassified as MSI-H by the artificial intelligence classifier. In the somatic MMR cohort, 11 (48%) had MSH6, 7 (30%) MLH1, 3 (13%) MSH2, and 2 (9%) PMS2. The median age at diagnosis was 72 years (range, 66-85 years). For composite MS status, 10 (43%) were MSI-H, 11 (48%) MS stable, and 2 (9%) MS indeterminate. Six cases were reclassified as MSI-H by the artificial intelligence classifier. For the LS and somatic MMR cohorts, 20 received ICB (n = 17 MSI-H). The median ICB duration was 27.7 months (95% CI, 11.5 to not reached); the disease control rate was 80%. Conclusion The results of this cross-sectional study suggest that MSI-H occurs due to LS or somatic oncogenesis in PC. Orthogonal MS testing is key in PC; the artificial intelligence classifier reclassified approximately 20% of cases, most of which were low cellularity. ICB for patients with LS or somatic MSI-H PC provided significant benefit.
Collapse
Affiliation(s)
- Catherine A. O’Connor
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Biology, Davidson College, Davidson, North Carolina
| | - Emily Harrold
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Mater Misericordiae University Hospital Dublin, Dublin, Ireland
| | - David Lin
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering, New York, New York
| | - Henry Walch
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering, New York, New York
| | - Andrea Gazzo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Megha Ranganathan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sarah Kane
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fergus Keane
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joshua Schoenfeld
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Drew Moss
- Mount Sinai Morningside West, New York, New York
| | | | - Sarah P. Suehnholz
- Human Oncology Pathogenesis Program, Sloan Kettering Institute, New York, New York
| | - Debyani Chakravarty
- Human Oncology Pathogenesis Program, Sloan Kettering Institute, New York, New York
| | - Fiyinfolu Balogun
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreas Cancer Research, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Anna Varghese
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreas Cancer Research, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Kenneth Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreas Cancer Research, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - David Kelsen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreas Cancer Research, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Alicia Latham
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreas Cancer Research, New York, New York
| | - Britta Weigelt
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Wungki Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreas Cancer Research, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Zsofia Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Eileen M. O’Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- David M. Rubenstein Center for Pancreas Cancer Research, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
17
|
Rodríguez-Olivares JL, Kimball TN, Jeter JM, De-La-Mora-Molina H, Núñez I, Weitzel JN, Chávarri-Guerra Y. Prevalence and spectrum of germline pathogenic variants in cancer susceptibility genes among mexican patients with exocrine pancreatic cancer. Pancreatology 2024; 24:1049-1056. [PMID: 39327123 DOI: 10.1016/j.pan.2024.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Although universal germline genetic testing is recommended for patients with exocrine pancreatic cancer (PC), access to genetic testing remains limited in low- and middle-income countries. This study aims to narrow the gap in our understanding of the spectrum of germline pathogenic and likely pathogenic variants (PVs) in cancer susceptibility genes in the Mexican population. METHODS The landscape of PVs in cancer susceptibility genes was identified by next-generation sequencing multigene panel assays among patients with PC who were enrolled in the Clinical Cancer Genomics Community Research Network prospective registry in Mexico City. RESULTS From August 2019 to April 2023, 137 patients underwent genetic testing. The median age at diagnosis was 60 years (range 36-85), 58.4 % were women, and 38.7 % were metastatic at diagnosis. The frequency of germline PVs was 16 % (n = 22): ATM 36.4 % (n = 8), CDKN2A/p16INK4A 27.3 % (n = 6), BRCA2 9.1 % (n = 2), PALB2 9.1 % (n = 2), CHEK2 9.1 % (n = 2), TP53 4.5 % (n = 1), and NF1 4.5 % (n = 1). Additionally, 2 carriers of monoallelic germline variants in MUTYH were identified. No significant differences were observed between carriers and non-carriers in terms of family history of pancreatic cancer. CONCLUSIONS We identified a significant frequency of actionable germline PVs in Mexicans with PC, wherein the majority were in a broad spectrum of genes associated with the homologous recombination DNA repair mechanism. Most pancreatic cancer associated PVs were detected in non-BRCA genes, so our findings support the recommendation of multigene panel testing for genetic cancer risk assessment of Mexican individuals with PC.
Collapse
Affiliation(s)
- José Luis Rodríguez-Olivares
- Department of Hematology and Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Tamara N Kimball
- Center for Genomic Medicine. Massachusetts General Hospital, Boston, MA, USA
| | - Joanne M Jeter
- Department of Oncology. City of Hope Cancer Center, Duarte, CA, USA
| | - Héctor De-La-Mora-Molina
- Department of Hematology and Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Isaac Núñez
- Research Division. Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jeffrey N Weitzel
- Division of Precision Prevention, University of Kansas Comprehensive Cancer Center, Kansas City, USA
| | - Yanin Chávarri-Guerra
- Department of Hematology and Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
18
|
Connelly CF, Towne WS, Desai N, Smithgall MC, Cimic A, Baskota SU. Cytologic testing for mismatch repair deficiency/microsatellite instability and NTRK gene fusion is not routinely indicated in primary pancreaticobiliary carcinoma cell block material. J Am Soc Cytopathol 2024; 13:413-419. [PMID: 39341739 DOI: 10.1016/j.jasc.2024.08.130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024]
Abstract
INTRODUCTION Pancreaticobiliary carcinomas rarely harbor targetable genetic alterations, including microsatellite instability (MSI) or neurotrophic tyrosine receptor kinase (NTRK) gene fusions. As these malignancies are typically present at an advanced stage and have suboptimal response to chemotherapy, the discovery of an actionable genomic alteration provides an additional avenue of treatment for chemotherapy-refractory cases. MATERIALS AND METHODS In this study, we evaluate 319 cases of pancreaticobiliary carcinoma diagnosed on fine-needle aspiration biopsy or biliary brushing for DNA mismatch repair (MMR) protein deficiency and pan-TRK overexpression by immunohistochemistry (IHC) and compare these results to MSI and NTRK gene fusion molecular testing. RESULTS AND CONCLUSION Although we find a high concordance between MMR protein IHC and MSI molecular testing in the evaluation of MMR deficiency and between pan-TRK IHC and NTRK fusion testing by next-generation sequencing, the low prevalence of either of these genetic alterations in our cohort casts doubt on the value of screening cases of pancreaticobiliary carcinoma for MMR protein deficiency and NTRK fusions.
Collapse
Affiliation(s)
- Courtney F Connelly
- Department of Pathology and Cell Biology, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York
| | - William S Towne
- Department of Pathology, St. Luke's University Health Network, Bethlehem, Pennsylvania
| | - Niyati Desai
- Department of Pathology and Cell Biology, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York
| | - Marie C Smithgall
- Department of Pathology and Cell Biology, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York
| | - Adela Cimic
- Department of Pathology and Cell Biology, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York
| | - Swikrity U Baskota
- Department of Pathology and Laboratory Medicine, University of California Irvine Health School of Medicine, Sacramento, California.
| |
Collapse
|
19
|
Hegazi A, Rager LE, Watkins DE, Su KH. Advancing Immunotherapy in Pancreatic Cancer. Int J Mol Sci 2024; 25:11560. [PMID: 39519112 PMCID: PMC11546161 DOI: 10.3390/ijms252111560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic cancer remains one of the deadliest malignancies, with a consistently low five-year survival rate for the past several decades. This is in stark contrast to other cancers, which have seen significant improvement in survival and prognosis due to recent developments in therapeutic modalities. These modest improvements in pancreatic cancer outcomes have primarily resulted from minor advances in cytotoxic chemotherapeutics, with limited progress in other treatment approaches. A major focus of current therapeutic research is the further development of immunomodulatory therapies characterized by antibody-based approaches, cellular therapies, and vaccines. Although initial results utilizing immunotherapy in pancreatic cancer have been mixed, recent clinical trials have demonstrated significant improvements in patient outcomes. In this review, we detail these three approaches to immunomodulation, highlighting their common targets and distinct shortcomings, and we provide a narrative summary of completed and ongoing clinical trials that utilize these approaches to immunomodulation. Within this context, we aim to inform future research efforts by identifying promising areas that warrant further exploration.
Collapse
Affiliation(s)
| | | | | | - Kuo-Hui Su
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; (A.H.); (L.E.R.); (D.E.W.)
| |
Collapse
|
20
|
Prezioso E, Mancheski E, Shivok K, Kaplan Z, Bowne W, Jain A, Lavu H, Yeo CJ, Nevler A. Assessing Influence of Mismatch Repair Mutations on Survival in Patients After Resection of Pancreatic Ductal and Periampullary Adenocarcinoma. J Clin Med 2024; 13:6185. [PMID: 39458133 PMCID: PMC11508431 DOI: 10.3390/jcm13206185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related deaths in the United States. Previous studies have indicated that microsatellite instability and deficient mismatch repair (MMR) may be associated with improved survival in patients with pancreatic cancer. Here, we aim to investigate the impact of deficient MMR (dMMR) status on oncologic outcomes in patients after resection of PDAC and periampullary adenocarcinoma. Methods: This is a single-institution, retrospective study based on a prospectively maintained database. Pancreatic ductal adenocarcinoma (N = 342) and periampullary adenocarcinoma patients (N = 76) who underwent pancreatic resection surgery between 2016 and 2021 were included. Immunohistochemistry staining results of MMR proteins and next-generation sequencing data were recorded. Cancer-type dependent Cox regression analyses were performed to assess overall and disease-free survival, which was complemented with a 1:2 propensity-score matching for each of the cancer types in order to compare oncologic outcomes. Results: A total of 418 pancreatic cancer patients were included in the analysis. Fifteen patients (3.5%) were diagnosed as dMMR (PDAC N = 7 and periampullary adenocarcinoma N = 8). Cox regression modeling of dMMR status interaction with TNM staging and cancer type revealed that dMMR status strongly improves overall survival (p < 0.05). After propensity-score matching, Cox regression identified dMMR status as a significant marker of improved overall survival (HR = 0.27, 95%CI 0.09-0.88, p = 0.029). Conclusions: Overall, our findings suggest that dMMR status is associated with markedly improved survival outcomes in patients after resection of pancreatic and periampullary cancer. Future large-scale studies are needed to further validate this finding.
Collapse
Affiliation(s)
| | | | - Kylee Shivok
- Sidney Kimmel Medical College, Philadelphia, PA 19107, USA; (E.P.)
| | - Zachary Kaplan
- Sidney Kimmel Medical College, Philadelphia, PA 19107, USA; (E.P.)
| | - Wilbur Bowne
- Jefferson Pancreatic, Biliary and Related Cancer Center, Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA
| | - Aditi Jain
- Jefferson Pancreatic, Biliary and Related Cancer Center, Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA
| | - Harish Lavu
- Jefferson Pancreatic, Biliary and Related Cancer Center, Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA
| | - Charles J. Yeo
- Jefferson Pancreatic, Biliary and Related Cancer Center, Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA
| | - Avinoam Nevler
- Jefferson Pancreatic, Biliary and Related Cancer Center, Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA
| |
Collapse
|
21
|
Piano MA, Boldrin E, Moserle L, Salerno N, Fanelli D, Peserico G, Biasin MR, Magni G, Varano V, Zalgelli G, Mourmouras V, Rosato A, Scapinello A, Fantin A, Curtarello M. MSI-H Detection by ddPCR in Endoscopic Ultrasound Fine Needle Biopsy (EUS-FNB) from Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2024; 25:11090. [PMID: 39456871 PMCID: PMC11507452 DOI: 10.3390/ijms252011090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive disease with limited survival. Curative opportunities are only available for patients with resectable cancer. Palliative chemotherapy is the current standard of care for unresectable tumors. Numerous efforts have been made to investigate new therapeutic strategies for PDAC. Immunotherapy has been found to be effective in treating tumors with high microsatellite instability (MSI-H), including PDAC. The ability of the Endoscopic Ultrasound Fine Needle Biopsy (EUS-FNB) to reliably collect tissue could enhance new personalized treatment by permitting genomic alterations analysis. The aim of this study was to investigate the feasibility of obtaining adequate DNA for molecular analysis from EUS-FNB formalin-fixed-paraffin-embedded (FFPE) specimens. For this purpose, FFPE-DNA obtained from 43 PDAC archival samples was evaluated to verify adequacy in terms of quantity and quality and was tested to evaluate MSI-H status by droplet digital PCR (ddPCR). All samples were suitable for ddPCR analysis. Unlike the 1-2% MSI-H frequency found with traditional techniques, ddPCR detected this phenotype in 16.28% of cases. This study suggests the ddPCR ability to identify MSI-H phenotype, with the possibility of improving the selection of patients who may benefit from immunotherapy and who would be excluded by performing traditional diagnostic methods.
Collapse
Affiliation(s)
- Maria Assunta Piano
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (M.A.P.); (E.B.); (L.M.); (N.S.); (V.V.); (A.R.)
| | - Elisa Boldrin
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (M.A.P.); (E.B.); (L.M.); (N.S.); (V.V.); (A.R.)
| | - Lidia Moserle
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (M.A.P.); (E.B.); (L.M.); (N.S.); (V.V.); (A.R.)
| | - Nicoletta Salerno
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (M.A.P.); (E.B.); (L.M.); (N.S.); (V.V.); (A.R.)
| | - Dalila Fanelli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Giulia Peserico
- Gastroenterology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (G.P.); (A.F.)
| | - Maria Raffaella Biasin
- Anatomy and Pathological Histology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (M.R.B.); (G.Z.); (V.M.); (A.S.)
| | - Giovanna Magni
- Clinical Research Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy;
| | - Veronica Varano
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (M.A.P.); (E.B.); (L.M.); (N.S.); (V.V.); (A.R.)
| | - Giorgia Zalgelli
- Anatomy and Pathological Histology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (M.R.B.); (G.Z.); (V.M.); (A.S.)
| | - Vasileios Mourmouras
- Anatomy and Pathological Histology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (M.R.B.); (G.Z.); (V.M.); (A.S.)
| | - Antonio Rosato
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (M.A.P.); (E.B.); (L.M.); (N.S.); (V.V.); (A.R.)
- Department of Surgery Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy
| | - Antonio Scapinello
- Anatomy and Pathological Histology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (M.R.B.); (G.Z.); (V.M.); (A.S.)
| | - Alberto Fantin
- Gastroenterology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (G.P.); (A.F.)
| | - Matteo Curtarello
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (M.A.P.); (E.B.); (L.M.); (N.S.); (V.V.); (A.R.)
| |
Collapse
|
22
|
Seufferlein T, Mayerle J, Boeck S, Brunner T, Ettrich TJ, Grenacher L, Gress TM, Hackert T, Heinemann V, Kestler A, Sinn M, Tannapfel A, Wedding U, Uhl W. S3-Leitlinie Exokrines Pankreaskarzinom – Version 3.1. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:874-995. [PMID: 39389103 DOI: 10.1055/a-2338-3533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Affiliation(s)
| | | | | | - Thomas Brunner
- Universitätsklinik für Strahlentherapie-Radioonkologie, Medizinische Universität Graz, Austria
| | | | | | - Thomas Mathias Gress
- Gastroenterologie und Endokrinologie Universitätsklinikum Gießen und Marburg, Germany
| | - Thilo Hackert
- Klinik und Poliklinik für Allgemein-, Viszeral- und Thoraxchirurgie, Universitätsklinikum Hamburg-Eppendorf, Germany
| | - Volker Heinemann
- Medizinische Klinik und Poliklinik III, Klinikum der Universität München-Campus Grosshadern, München, Germany
| | | | - Marianne Sinn
- Medizinische Klinik und Poliklinik II Onkologie und Hämatologie, Universitätsklinikum Hamburg-Eppendorf, Germany
| | | | | | - Waldemar Uhl
- Allgemein- und Viszeralchirurgie, St Josef-Hospital, Bochum, Germany
| |
Collapse
|
23
|
Brugiapaglia S, Spagnolo F, Intonti S, Novelli F, Curcio C. Fighting Pancreatic Cancer with a Vaccine-Based Winning Combination: Hope or Reality? Cells 2024; 13:1558. [PMID: 39329742 PMCID: PMC11430323 DOI: 10.3390/cells13181558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/06/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024] Open
Abstract
Pancreatic adenocarcinoma (PDA) represents the fourth leading cause of cancer-related mortality in the USA. Only 20% of patients present surgically resectable and potentially curable tumors at diagnosis, while 80% are destined for poor survival and palliative chemotherapy. Accordingly, the advancement of innovative and effective therapeutic strategies represents a pivotal medical imperative. It has been demonstrated that targeting the immune system represents an effective approach against several solid tumors. The immunotherapy approach encompasses a range of strategies, including the administration of antibodies targeting checkpoint molecules (immune checkpoint inhibitors, ICIs) to disrupt tumor suppression mechanisms and active immunization approaches that aim to stimulate the host's immune system. While vaccines have proved effective against infectious agents, vaccines for cancer remain an unfulfilled promise. Vaccine-based therapy targeting tumor antigens has the potential to be a highly effective strategy for initiating and maintaining T cell recognition, enhancing the immune response, and ultimately promoting cancer treatment success. In this review, we examined the most recent clinical trials that employed diverse vaccine types to stimulate PDA patients' immune systems, either independently or in combination with chemotherapy, radiotherapy, ICIs, and monoclonal antibodies with the aim of ameliorating PDA patients' quality of life and extend their survival.
Collapse
Affiliation(s)
- Silvia Brugiapaglia
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44bis, 10126 Turin, Italy; (S.B.); (S.I.); (F.N.)
| | - Ferdinando Spagnolo
- School of Advanced Defence Studies, Defence Research & Analysis Institute, Piazza della Rovere 83, 00165 Rome, Italy; (F.S.)
| | - Simona Intonti
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44bis, 10126 Turin, Italy; (S.B.); (S.I.); (F.N.)
| | - Francesco Novelli
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44bis, 10126 Turin, Italy; (S.B.); (S.I.); (F.N.)
| | - Claudia Curcio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44bis, 10126 Turin, Italy; (S.B.); (S.I.); (F.N.)
| |
Collapse
|
24
|
Netto D, Frizziero M, Foy V, McNamara MG, Backen A, Hubner RA. Systemic Therapy for Metastatic Pancreatic Cancer-Current Landscape and Future Directions. Curr Oncol 2024; 31:5206-5223. [PMID: 39330013 PMCID: PMC11430697 DOI: 10.3390/curroncol31090385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 09/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a significant cause of cancer-associated mortality, with a rising global incidence. A paucity of strong predictive risk factors mean screening programmes are difficult to implement. Historically, a lack of identifiable and actionable driver mutations, coupled with a relatively immunosuppressed tumour microenvironment, has led to a reliance on cytotoxic chemotherapy. The NAPOLI-3 trial has reported data supporting consideration of NALIRIFOX as a new first-line standard of care. Kirsten Rat Sarcoma Virus (KRAS) G12D mutations are present in >90% of all PDAC's; exciting breakthroughs in small molecule inhibitors targeting KRAS G12D may open new modalities of treatment, and therapies targeting multiple KRAS mutations are also in early clinical trials. Although immunotherapy strategies to date have been disappointing, combination with chemotherapy and/or small molecule inhibitors hold promise and warrant further exploration.
Collapse
Affiliation(s)
- Daniel Netto
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Melissa Frizziero
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Victoria Foy
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Mairéad G. McNamara
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Alison Backen
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Richard A. Hubner
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| |
Collapse
|
25
|
Kwan MC, Zhang ML. Pancreas Fine Needle Aspiration: Current and Future Impact on Patient Care. Surg Pathol Clin 2024; 17:441-452. [PMID: 39129142 DOI: 10.1016/j.path.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Pancreatic lesions can be solid or cystic and comprise a wide range of benign, premalignant, and malignant entities. Endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) is the current primary sampling method for the preoperative diagnosis of pancreatic lesions. Optimal handling of cytology/small tissue specimens is critical to ensure that the often-scant diagnostic material is appropriately utilized for ancillary and/or molecular studies when appropriate. Ultimately, evaluation of EUS-FNA cytology and small biopsy material can provide accurate and timely diagnoses to guide patient management and triage them to surveillance or surgical intervention.
Collapse
Affiliation(s)
- Melanie C Kwan
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA. https://twitter.com/melaniekwan
| | - M Lisa Zhang
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Merz V, Maines F, Marcucci S, Sartori C, Frisinghelli M, Trentin C, Kadrija D, Carbone FG, Michielan A, Gabbrielli A, Melisi D, Barbareschi M, Brolese A, Caffo O. Complete pathological response to pembrolizumab in pretreated pancreatic acinar cell carcinoma. J Cancer Res Clin Oncol 2024; 150:347. [PMID: 38990367 PMCID: PMC11239721 DOI: 10.1007/s00432-024-05841-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/06/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Therapeutic approach used for pancreatic ductal adenocarcinoma is usually translated also for the rarer acinar counterpart, which shows a different mutational landscape nevertheless. While dMMR/MSI-H status is rare in the ductal histotype, it appears to be more prevalent in pancreatic acinar cell carcinoma (PACC). CASE PRESENTATION We report the case of a patient with locally advanced MSI-H PACC in whom the treatment with the anti-PD-1 pembrolizumab, administered as third line, made possible surgical resection, achieving even an exceptional pathological complete response. CONCLUSIONS Treatment of PACC should be tailored based on the peculiar molecular features that distinguish PACC from ductal adenocarcinoma. Evaluation of potentially therapeutically targetable alterations should be mandatory in case of PACC diagnosis.
Collapse
Affiliation(s)
- Valeria Merz
- Department of Medical Oncology, Santa Chiara Hospital, APSS, L.Go Medaglie d'Oro,9, 38122, Trento, Italy.
- Digestive Molecular Clinical Oncology Research Unit, Università degli Studi di Verona, Verona, Italy.
| | - Francesca Maines
- Department of Medical Oncology, Santa Chiara Hospital, APSS, L.Go Medaglie d'Oro,9, 38122, Trento, Italy
| | - Stefano Marcucci
- Department of General Surgery and HPB Unit, Santa Chiara Hospital, APSS, Trento, Italy
| | - Chiara Sartori
- Department of Laboratory Medicine - Pathology Unit, Santa Chiara Hospital, APSS, Trento, Italy
| | - Michela Frisinghelli
- Department of Medical Oncology, Santa Chiara Hospital, APSS, L.Go Medaglie d'Oro,9, 38122, Trento, Italy
| | - Chiara Trentin
- Department of Medical Oncology, Santa Chiara Hospital, APSS, L.Go Medaglie d'Oro,9, 38122, Trento, Italy
| | - Dzenete Kadrija
- Department of Medical Oncology, Santa Chiara Hospital, APSS, L.Go Medaglie d'Oro,9, 38122, Trento, Italy
| | | | - Andrea Michielan
- Gastroenterology and Digestive Endoscopy Unit, Santa Chiara Hospital, APSS, Trento, Italy
| | - Armando Gabbrielli
- Gastroenterology and Digestive Endoscopy Unit, Santa Chiara Hospital, APSS, Trento, Italy
- Center for Medical Sciences (CISMed), University of Trento, Trento, Italy
| | - Davide Melisi
- Digestive Molecular Clinical Oncology Research Unit, Università degli Studi di Verona, Verona, Italy
| | - Mattia Barbareschi
- Department of Laboratory Medicine - Pathology Unit, Santa Chiara Hospital, APSS, Trento, Italy
- Center for Medical Sciences (CISMed), University of Trento, Trento, Italy
| | - Alberto Brolese
- Department of General Surgery and HPB Unit, Santa Chiara Hospital, APSS, Trento, Italy
| | - Orazio Caffo
- Department of Medical Oncology, Santa Chiara Hospital, APSS, L.Go Medaglie d'Oro,9, 38122, Trento, Italy
| |
Collapse
|
27
|
Tezuka K, Yamakawa M, Murakami R, Hirai I, Toya R, Suzuki A, Kawamura H, Miyano Y, Sato H, Motoi F. Familial Intraductal Papillary Mucinous Neoplasm Associated With the Germline MSH6 Missense Variant and Progression of Pancreatic cancer. Pancreas 2024; 53:e476-e486. [PMID: 38416847 DOI: 10.1097/mpa.0000000000002313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/01/2024]
Abstract
OBJECTIVES Intraductal papillary mucinous neoplasm (IPMN) in individuals with at least one first-degree relative with IPMN is defined as familial IPMN. However, few studies have reported on familial IPMN, its clinical characteristics, or the associated genetic factors. MATERIALS AND METHODS We report the case of a 58-year-old woman with multifocal IPMN and a mural nodule in the pancreatic body. The patient underwent a distal pancreatectomy and developed pancreatic head cancer 1 year and 6 months postoperatively. The patient had a family history of multifocal IPMN in her father. Therefore, a genetic predisposition to IPMN and pancreatic cancer was suspected. The patient was analyzed for germline variants, and the resected IPMN was subjected to immunohistochemical and somatic variant analyses. RESULTS Next-generation sequencing revealed a heterozygous germline missense variant in exon 5 of MSH6 (c.3197A>G; Tyr1066Cys). The pathogenicity of this variant of uncertain significance was suspected based on multiple in silico analyses, and the same MSH6 variant was identified in the patient's father's colonic adenoma. The mural nodule in the pancreatic body was pathologically diagnosed as a high-grade IPMN with ossification and somatic KRAS and PIK3CA variants. CONCLUSIONS This case revealed a possible genetic factor for familial IPMN development and presented interesting clinicopathological findings.
Collapse
Affiliation(s)
| | | | - Ryoko Murakami
- Genome Informatics Unit, Institution for Promotion of Medical Science Research, Yamagata University Faculty of Medicine
| | | | | | | | | | - Yuki Miyano
- Genome Informatics Unit, Institution for Promotion of Medical Science Research, Yamagata University Faculty of Medicine
| | - Hidenori Sato
- Genome Informatics Unit, Institution for Promotion of Medical Science Research, Yamagata University Faculty of Medicine
| | - Fuyuhiko Motoi
- First Department of Surgery, Yamagata University Faculty of Medicine, Yamagata, Japan
| |
Collapse
|
28
|
Mencel J, Alves A, Angelis V, Gerlinger M, Starling N. State of the art: Targeting microsatellite instability in gastrointestinal cancers. Crit Rev Oncol Hematol 2024; 199:104387. [PMID: 38734279 DOI: 10.1016/j.critrevonc.2024.104387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
DNA mismatch repair (MMR) deficiency and the associated microsatellite instability (MSI) phenotype has become a subject of enormous interest in recent years due to the demonstrated efficacy of immune checkpoint inhibitors (ICI) in advanced tumours. Assessing MSI in patients with gastrointestinal tract (GI) cancers is useful to exclude Lynch syndrome, but also to predict benefit for ICI. Following review of the relevant literature, this review article aims to outline the clinicopathologic spectrum of MSI and mismatch repair deficiency (dMMR) in the GI tract, hepatobiliary system and pancreas and discuss the therapeutic consideration in this disease.
Collapse
Affiliation(s)
- Justin Mencel
- Gastrointestinal and Lymphoma Unit, Royal Marsden NHS Foundation, London, United Kingdom
| | - Anneke Alves
- Gastrointestinal and Lymphoma Unit, Royal Marsden NHS Foundation, London, United Kingdom
| | - Vasileios Angelis
- Gastrointestinal and Lymphoma Unit, Royal Marsden NHS Foundation, London, United Kingdom
| | - Marco Gerlinger
- Gastrointestinal and Lymphoma Unit, Royal Marsden NHS Foundation, London, United Kingdom
| | - Naureen Starling
- Gastrointestinal and Lymphoma Unit, Royal Marsden NHS Foundation, London, United Kingdom.
| |
Collapse
|
29
|
Cortiana V, Abbas RH, Chorya H, Gambill J, Mahendru D, Park CH, Leyfman Y. Personalized Medicine in Pancreatic Cancer: The Promise of Biomarkers and Molecular Targeting with Dr. Michael J. Pishvaian. Cancers (Basel) 2024; 16:2329. [PMID: 39001391 PMCID: PMC11240738 DOI: 10.3390/cancers16132329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Pancreatic cancer, with its alarming rising incidence, is predicted to become the second deadliest type of solid tumor by 2040, highlighting the urgent need for improved diagnostic and treatment strategies. Despite medical advancements, the five-year survival rate for pancreatic cancer remains about 14%, dropping further when metastasized. This review explores the promise of biomarkers for early detection, personalized treatment, and disease monitoring. Molecular classification of pancreatic cancer into subtypes based on genetic mutations, gene expression, and protein markers guides treatment decisions, potentially improving outcomes. A plethora of clinical trials investigating different strategies are currently ongoing. Targeted therapies, among which those against CLAUDIN 18.2 and inhibitors of Claudin 18.1, have shown promise. Next-generation sequencing (NGS) has emerged as a powerful tool for the comprehensive genomic analysis of pancreatic tumors, revealing unique genetic alterations that drive cancer progression. This allows oncologists to tailor therapies to target specific molecular abnormalities. However, challenges remain, including limited awareness and uptake of biomarker-guided therapies. Continued research into the molecular mechanisms of pancreatic cancer is essential for developing more effective treatments and improving patient survival rates.
Collapse
Affiliation(s)
- Viviana Cortiana
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | | | | | | | - Diksha Mahendru
- Global Remote Research Scholars Program, St. Paul, MN 55101, USA
| | | | - Yan Leyfman
- Icahn School of Medicine at Mount Sinai South Nassau, Oceanside, NY 11572, USA
| |
Collapse
|
30
|
Chen S, Xie D, Li Z, Wang J, Hu Z, Zhou D. Frequency-dependent selection of neoantigens fosters tumor immune escape and predicts immunotherapy response. Commun Biol 2024; 7:770. [PMID: 38918569 PMCID: PMC11199503 DOI: 10.1038/s42003-024-06460-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Cancer is an evolutionary process shaped by selective pressure from the microenvironments. However, recent studies reveal that certain tumors undergo neutral evolution where there is no detectable fitness difference amongst the cells following malignant transformation. Here, through computational modeling, we demonstrate that negative frequency-dependent selection (or NFDS), where the immune response against cancer cells depends on the clonality of neoantigens, can lead to an immunogenic landscape that is highly similar to neutral evolution. Crucially, NFDS promotes high antigenic heterogeneity and early immune evasion in hypermutable tumors, leading to poor responses to immune checkpoint blockade (ICB) therapy. Our model also reveals that NFDS is characterized by a negative association between average clonality and total burden of neoantigens. Indeed, this unique feature of NFDS is common in the whole-exome sequencing (WES) datasets (357 tumor samples from 275 patients) from four melanoma cohorts with ICB therapy and a non-small cell lung cancer (NSCLC) WES dataset (327 tumor samples from 100 patients). Altogether, our study provides quantitative evidence supporting the theory of NFDS in cancer, explaining the high prevalence of neutral-looking tumors. These findings also highlight the critical role of frequency-dependent selection in devising more efficient and predictive immunotherapies.
Collapse
Affiliation(s)
- Shaoqing Chen
- School of Mathematical Sciences, Xiamen University, Xiamen, China
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China
| | - Duo Xie
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Zan Li
- Life Science Research Center, Core Research Facilities, Southern University of Science and Technology, Shenzhen, China
| | - Jiguang Wang
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
- Hong Kong Center for Neurodegenerative Diseases, InnoHK, Hong Kong SAR, China
- HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen, China
| | - Zheng Hu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Da Zhou
- School of Mathematical Sciences, Xiamen University, Xiamen, China.
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
31
|
Kuznetsova AV, Glukhova XA, Popova OP, Beletsky IP, Ivanov AA. Contemporary Approaches to Immunotherapy of Solid Tumors. Cancers (Basel) 2024; 16:2270. [PMID: 38927974 PMCID: PMC11201544 DOI: 10.3390/cancers16122270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/11/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
In recent years, the arrival of the immunotherapy industry has introduced the possibility of providing transformative, durable, and potentially curative outcomes for various forms of malignancies. However, further research has shown that there are a number of issues that significantly reduce the effectiveness of immunotherapy, especially in solid tumors. First of all, these problems are related to the protective mechanisms of the tumor and its microenvironment. Currently, major efforts are focused on overcoming protective mechanisms by using different adoptive cell therapy variants and modifications of genetically engineered constructs. In addition, a complex workforce is required to develop and implement these treatments. To overcome these significant challenges, innovative strategies and approaches are necessary to engineer more powerful variations of immunotherapy with improved antitumor activity and decreased toxicity. In this review, we discuss recent innovations in immunotherapy aimed at improving clinical efficacy in solid tumors, as well as strategies to overcome the limitations of various immunotherapies.
Collapse
Affiliation(s)
- Alla V. Kuznetsova
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia
| | - Xenia A. Glukhova
- Onni Biotechnologies Ltd., Aalto University Campus, Metallimiehenkuja 10, 02150 Espoo, Finland; (X.A.G.); (I.P.B.)
| | - Olga P. Popova
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
| | - Igor P. Beletsky
- Onni Biotechnologies Ltd., Aalto University Campus, Metallimiehenkuja 10, 02150 Espoo, Finland; (X.A.G.); (I.P.B.)
| | - Alexey A. Ivanov
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
| |
Collapse
|
32
|
Emelyanova M, Ikonnikova A, Pushkov A, Pudova E, Krasnov G, Popova A, Zhanin I, Khomich D, Abramov I, Tjulandin S, Gryadunov D, Pokataev I. Mutations in Mismatch Repair Genes and Microsatellite Instability Status in Pancreatic Cancer. Cancers (Basel) 2024; 16:2111. [PMID: 38893230 PMCID: PMC11171205 DOI: 10.3390/cancers16112111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/19/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Patients with pancreatic cancer (PC) showing mismatch repair (MMR) deficiency may benefit from immunotherapy. Microsatellite instability (MSI) is a hallmark of MMR deficiency (MMR-D). Here, we estimated the prevalence of MSI in PC, investigated germline and somatic mutations in the three MMR genes (MLH1, MSH2, and MSH6), and assessed the relationship between MMR genes mutations and MSI status in PC. Clinical specimens from PC patients were analyzed using targeted next-generation sequencing, including paired normal and tumor specimens from 155 patients, tumor-only specimens from 86 patients, and normal-only specimens from 379 patients. The MSI status of 235 PCs was assessed via PCR. Pathogenic/likely pathogenic (P/LP) germline variants in the MMR genes were identified in 1.1% of patients, while somatic variants were found in 2.6% of patients. No MSI-H tumors were detected. One patient carried two variants (P (VAF = 0.57) and LP (VAF = 0.25)) simultaneously; however, their germline/somatic status remains unknown due to the investigation focusing solely on the tumor and MSI analysis was not performed for this patient. MSI is rare in PC, even in tumors with MMR genes mutations. Our findings underscore the importance of assessing tumor MMR-D status in PC patients with confirmed Lynch syndrome when deciding whether to prescribe immunotherapy.
Collapse
Affiliation(s)
- Marina Emelyanova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; (A.I.); (E.P.); (G.K.); (D.K.); (I.A.); (D.G.)
| | - Anna Ikonnikova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; (A.I.); (E.P.); (G.K.); (D.K.); (I.A.); (D.G.)
| | - Alexander Pushkov
- Federal State Autonomous Institution “National Medical Research Center for Children’s Health” of the Ministry of Health of the Russian Federation, Moscow 119991, Russia; (A.P.); (I.Z.)
| | - Elena Pudova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; (A.I.); (E.P.); (G.K.); (D.K.); (I.A.); (D.G.)
| | - George Krasnov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; (A.I.); (E.P.); (G.K.); (D.K.); (I.A.); (D.G.)
| | - Anna Popova
- N.N. Blokhin National Medical Research Center for Oncology, Ministry of Health of the Russian Federation, Moscow 115522, Russia; (A.P.); (S.T.); (I.P.)
| | - Ilya Zhanin
- Federal State Autonomous Institution “National Medical Research Center for Children’s Health” of the Ministry of Health of the Russian Federation, Moscow 119991, Russia; (A.P.); (I.Z.)
| | - Darya Khomich
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; (A.I.); (E.P.); (G.K.); (D.K.); (I.A.); (D.G.)
| | - Ivan Abramov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; (A.I.); (E.P.); (G.K.); (D.K.); (I.A.); (D.G.)
| | - Sergei Tjulandin
- N.N. Blokhin National Medical Research Center for Oncology, Ministry of Health of the Russian Federation, Moscow 115522, Russia; (A.P.); (S.T.); (I.P.)
| | - Dmitry Gryadunov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; (A.I.); (E.P.); (G.K.); (D.K.); (I.A.); (D.G.)
| | - Ilya Pokataev
- N.N. Blokhin National Medical Research Center for Oncology, Ministry of Health of the Russian Federation, Moscow 115522, Russia; (A.P.); (S.T.); (I.P.)
- City Clinical Cancer Hospital No 1, Moscow Department of Health, Moscow 129090, Russia
| |
Collapse
|
33
|
Limijadi EKS, Muniroh M, Prajoko YW, Tjandra KC, Respati DRP. The role of germline BRCA1 & BRCA2 mutations in familial pancreatic cancer: A systematic review and meta-analysis. PLoS One 2024; 19:e0299276. [PMID: 38809921 PMCID: PMC11135687 DOI: 10.1371/journal.pone.0299276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/15/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Familial Pancreatic Cancer (FPC) presents a notable risk, with 3-10% of pancreatic adenocarcinoma cases having a family history. Studies link FPC to syndromes like HBOC, suggesting BRCA1/BRCA2 mutations play a role. BRCA gene functions in DNA repair impact FPC management, influencing sensitivity to therapies like PARP inhibitors. Identifying mutations not only aids FPC treatment but also reveals broader cancer risks. However, challenges persist in selectively applying genetic testing due to cost constraints. This Systematic Review focuses on BRCA1/BRCA2 significance in FPC, diagnostic criteria, prognostic value, and limitations. METHOD Original articles published from 2013 to January 2023 were sourced from databases such as Scopus, PubMed, ProQuest, and ScienceDirect. Inclusion criteria comprised observational cohort or diagnostic studies related to the role of BRCA1/2 mutation in correlation to familial pancreatic cancer (FPC), while article reviews, narrative reviews, and non-relevant content were excluded. The assessment of bias used ROBINS-I, and the results were organized using PICOS criteria in a Google spreadsheet table. The systematic review adhered to the PRISMA 2020 checklist. RESULT We analyzed 9 diagnostic studies encompassing 1325 families and 4267 patients from Italy, USA, and Poland. Despite the limitation of limited homogenous PICO studies, our findings effectively present evidence. BRCA1/2 demonstrates benefits in detecting first-degree relatives FPC involvement with 2.26-10 times higher risk. These mutation findings also play an important role since with the BRCA1/2 targeted therapy, Poly-ADP Ribose Polymerase inhibitors (PARP) may give better outcomes of FPC treatment. Analysis of BRCA1 and BRCA2 administration's impact on odds ratio (OR) based on six and five studies respectively. BRCA1 exhibited non-significant effects (OR = 1.26, P = 0.51), while BRCA2 showed significance (OR = 1.68, P = 0.04). No heterogeneity observed, indicating consistent results. Further research on BRCA1 is warranted. CONCLUSION Detecting the BRCA1/2 mutation gene offers numerous advantages, particularly in its correlation with FPC. For diagnostic and prognostic purposes, testing is strongly recommended for first-degree relatives, who face a significantly higher risk (2.26-10 times) of being affected. Additionally, FPC patients with identified BRCA1/2 mutations exhibit a more favorable prognosis compared to the non-mutated population. This is attributed to the availability of targeted BRCA1/2 therapy, which maximizes treatment outcomes.
Collapse
Affiliation(s)
- Edward Kurnia Setiawan Limijadi
- Doctoral Study Program of Medical and Health Science, Universitas Diponegoro, Semarang, Indonesia
- Faculty of Medicine, Department of Clinical Pathology, Universitas Diopnegoro, Semarang, Indonesia
| | - Muflihatul Muniroh
- Faculty of Medicine, Department of Physiology, Universitas Diponegoro, Semarang, Indonesia
| | - Yan Wisnu Prajoko
- Faculty of Medicine, Department of Surgical Oncology, Universitas Diponegoro, Semarang, Indonesia
- Kariadi General Hospital, Semarang, Indonesia
| | - Kevin Christian Tjandra
- Kariadi General Hospital, Semarang, Indonesia
- Faculty of Medicine, Departement of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Danendra Rakha Putra Respati
- Kariadi General Hospital, Semarang, Indonesia
- Faculty of Medicine, Departement of Medicine, Universitas Diponegoro, Semarang, Indonesia
| |
Collapse
|
34
|
Desai A, Xiao AH, Choi D, Toruner MD, Walden D, Halfdanarson TR, Alberts S, McWilliams RR, Mahipal A, Ahn D, Babiker H, Stybayeva G, Revzin A, Kizilbash S, Adjei A, Bekaii-Saab T, Mansfield AS, Carr RM, Ma WW. Molecular Characterization and Therapeutic Opportunities in KRAS Wildtype Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:1861. [PMID: 38791940 PMCID: PMC11119482 DOI: 10.3390/cancers16101861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
PURPOSE To investigate the molecular characteristics of and potential for precision medicine in KRAS wildtype pancreatic ductal adenocarcinoma (PDAC). PATIENTS AND METHODS We investigated 27 patients with KRASWT PDAC at our institution. Clinical data were obtained via chart review. Tumor specimens for each subject were interrogated for somatic single nucleotide variants, insertion and deletions, and copy number variants by DNA sequencing. Gene fusions were detected from RNA-seq. A patient-derived organoid (PDO) was developed from a patient with a MET translocation and expanded ex vivo to predict therapeutic sensitivity prior to enrollment in a phase 2 clinical trial. RESULTS Transcriptomic analysis showed our cohort may be stratified by the relative gene expression of the KRAS signaling cascade. The PDO derived from our patient harboring a TFG-MET rearrangement was found to have in vitro sensitivity to the multi-tyrosine kinase inhibitor crizotinib. The patient was enrolled in the phase 2 SPARTA clinical trial and received monotherapy with vebrelitinib, a c-MET inhibitor, and achieved a partial and durable response. CONCLUSIONS KRASWT PDAC is molecularly distinct from KRASMUT and enriched with potentially actionable genetic variants. In our study, transcriptomic profiling revealed that the KRAS signaling cascade may play a key role in KRASWT PDAC. Our report of a KRASWT PDAC patient with TFG-MET rearrangement who responded to a cMET inhibitor further supports the pursuit of precision oncology in this sub-population. Identification of targetable mutations, perhaps through approaches like RNA-seq, can help enable precision-driven approaches to select optimal treatment based on tumor characteristics.
Collapse
Affiliation(s)
- Aakash Desai
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | | | - Daheui Choi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55902, USA; (D.C.); (G.S.); (A.R.)
| | - Merih D. Toruner
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | - Daniel Walden
- Division of Hematology and Oncology, Mayo Clinic, Phoenix, AZ 85054, USA; (D.W.); (D.A.); (T.B.-S.)
| | - Thorvardur R. Halfdanarson
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | - Steven Alberts
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | - Robert R. McWilliams
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | - Amit Mahipal
- Department of Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA;
| | - Daniel Ahn
- Division of Hematology and Oncology, Mayo Clinic, Phoenix, AZ 85054, USA; (D.W.); (D.A.); (T.B.-S.)
| | - Hani Babiker
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55902, USA; (D.C.); (G.S.); (A.R.)
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55902, USA; (D.C.); (G.S.); (A.R.)
| | - Sani Kizilbash
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | - Alex Adjei
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Tanios Bekaii-Saab
- Division of Hematology and Oncology, Mayo Clinic, Phoenix, AZ 85054, USA; (D.W.); (D.A.); (T.B.-S.)
| | - Aaron S. Mansfield
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | - Ryan M. Carr
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | - Wen Wee Ma
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| |
Collapse
|
35
|
Ward JD, Fowler M, Robledo-Gomez A, Goodyear SM, Kardosh A, Sasatomi E. PD-L1 expression in pancreaticobiliary adenosquamous carcinoma: a single-institution case series. J Gastrointest Oncol 2024; 15:768-779. [PMID: 38756636 PMCID: PMC11094501 DOI: 10.21037/jgo-24-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/31/2024] [Indexed: 05/18/2024] Open
Abstract
Background The programmed cell death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1) pathway is a potent negative regulator of T-cell-mediated immune response that is upregulated in many neoplasms. Pancreaticobiliary adenosquamous carcinoma (PB-ASC) is an aggressive cancer that carries a poorer prognosis compared with pure pancreaticobiliary adenocarcinoma (PB-AC). To date, there is little published information regarding PD-L1 expression in PB-ASC. The aim of the study was to examine the relationship between PD-L1 expression and tumor-infiltrating lymphocytes in PB-ASC and PB-AC. Methods We evaluated 15 PB-ASCs (10 pancreatic, 5 gallbladder) and 34 control PB-ACs (22 pancreatic ductal, and 12 gallbladder) for tumor expression of PD-L1 using anti-PD-L1 (E1L3N) antibody. All tumors were classified into three immune phenotypes: immune inflamed (II), immune excluded (IE), and immune desert (ID) according to the distribution of tumor-infiltrating lymphocytes in tumor tissues. Results The frequency of PD-L1 expression was significantly higher in PB-ASC (10/15; 66.7%) than in PB-AC (3/34; 8.8%). In PB-ASC, PD-L1 expression occurred exclusively in the squamous component in six cases, exclusively in the glandular component in one case, and in both the squamous and the glandular components in three cases. PD-L1 expression in PB-ASC was irrespective of the tumor immune status, whereas its expression in PB-AC was observed only in tumors with the II or IE phenotype. The ID phenotype was relatively rare (4/15; 26.7%) in PB-ASC compared with PB-AC (22/34; 65%; P=0.02). Conclusions PB-ASCs are notably enriched in inflammatory response and showed significantly higher PD-L1 expression than PB-AC (P<0.001), suggesting a potential therapeutic role for immune checkpoint inhibitors in managing patients with PB-ASC.
Collapse
Affiliation(s)
- Jeremy D. Ward
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Mark Fowler
- Department of Pathology, Community Memorial Hospital, Ventura, CA, USA
| | - Ariannette Robledo-Gomez
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University (OHSU), Portland, OR, USA
| | - Shaun M. Goodyear
- Knight Cancer Institute, OHSU, Portland, OR, USA
- Division of Hematology and Oncology, School of Medicine, OHSU, Portland, OR, USA
| | - Adel Kardosh
- Knight Cancer Institute, OHSU, Portland, OR, USA
- Division of Hematology and Oncology, School of Medicine, OHSU, Portland, OR, USA
| | - Eizaburo Sasatomi
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University (OHSU), Portland, OR, USA
| |
Collapse
|
36
|
Skorupan N, Peer CJ, Zhang X, Choo-Wosoba H, Ahmad MI, Lee MJ, Rastogi S, Sato N, Yu Y, Pegna GJ, Steinberg SM, Kalsi SS, Cao L, Figg WD, Trepel JB, Pastan I, FitzGerald D, Alewine C. Tofacitinib to prevent anti-drug antibody formation against LMB-100 immunotoxin in patients with advanced mesothelin-expressing cancers. Front Oncol 2024; 14:1386190. [PMID: 38706610 PMCID: PMC11066227 DOI: 10.3389/fonc.2024.1386190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/22/2024] [Indexed: 05/07/2024] Open
Abstract
Background LMB-100 is a mesothelin (MSLN)-targeting recombinant immunotoxin (iTox) carrying a Pseudomonas exotoxin A payload that has shown promise against solid tumors, however, efficacy is limited by the development of neutralizing anti-drug antibodies (ADAs). Tofacitinib is an oral Janus Kinase (JAK) inhibitor that prevented ADA formation against iTox in preclinical studies. Methods A phase 1 trial testing LMB-100 and tofacitinib in patients with MSLN-expressing cancers (pancreatic adenocarcinoma, n=13; cholangiocarcinoma, n=1; appendiceal carcinoma, n=1; cystadenocarcinoma, n=1) was performed to assess safety and to determine if tofacitinib impacted ADA formation. Participants were treated for up to 3 cycles with LMB-100 as a 30-minute infusion on days 4, 6, and 8 at two dose levels (100 and 140 µg/kg) while oral tofacitinib was administered for the first 10 days of the cycle (10 mg BID). Peripheral blood was collected for analysis of ADA levels, serum cytokines and circulating immune subsets. Results The study was closed early due to occurrence of drug-induced pericarditis in 2 patients. Pericarditis with the combination was not reproducible in a transgenic murine model containing human MSLN. Two of 4 patients receiving all 3 cycles of treatment maintained effective LMB-100 levels, an unusual occurrence. Sustained increases in systemic IL-10 and TNF-α were seen, a phenomenon not observed in prior LMB-100 studies. A decrease in activated T cell subsets and an increase in circulating immunosuppressive myeloid populations occurred. No radiologic decreases in tumor volume were observed. Discussion Further testing of tofacitinib to prevent ADA formation is recommended in applicable non-malignant disease settings. Clinical trial registration https://www.clinicaltrials.gov/study/NCT04034238.
Collapse
Affiliation(s)
- Nebojsa Skorupan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Cody J. Peer
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Xianyu Zhang
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Hyoyoung Choo-Wosoba
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mehwish I. Ahmad
- Office of Research Nursing, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Min-Jung Lee
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Shraddha Rastogi
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Nahoko Sato
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yunkai Yu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Guillaume Joe Pegna
- Medical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Shelley S. Kalsi
- Hematology Consult and Graduate Medical Section, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Liang Cao
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - William D. Figg
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jane B. Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - David FitzGerald
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Christine Alewine
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
37
|
Notohara K, Nakamura K. Tissue processing of endoscopic ultrasound-guided fine-needle aspiration specimens from solid pancreatic lesions. J Med Ultrason (2001) 2024; 51:261-274. [PMID: 38051462 DOI: 10.1007/s10396-023-01387-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/04/2023] [Indexed: 12/07/2023]
Abstract
Now that tissue cores can be obtained using fine-needle biopsy (FNB) needles, the ways tissues are handled for endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA) are changing. Direct smear, touch smear of core tissues, and centrifugation have been used for cytological examinations, and liquid-based cytology (LBC), which allows immunostaining and genetic tests that use residual samples, is emerging as an alternative. We emphasize that liquid cytology (Cytospin™ cytology and LBC) is still important, because it enables the diagnosis of pancreatic ductal adenocarcinoma (PDAC) when cancerous cells are scarce in specimens. Cell blocks are being replaced by core tissues obtained via FNB needles. Recent reports indicate that rapid on-site evaluation (ROSE) is not necessary when FNB needles are used, and macroscopic on-site evaluation is used to evaluate specimen adequacy. Macroscopic findings of specimens are helpful in the diagnostic workup and for clarifying specimen-handling methods. In addition to the red strings and white cores observed in PDAC, mixed red and white strings, gray tissues, and gelatinous tissues are observed. Gray (necrotic) tissues and gelatinous (mucus) tissues are more suitable than histology for cell block or cytological processing. Tumor cells in neuroendocrine tumors (NETs) are numerous in red strings but cannot be observed macroscopically. ROSE might thus be necessary for lesions that may be NETs. Core tissues can be used for genetic tests, such as those used for KRAS mutations and comprehensive genomic profiling. Cytological materials, including slides and LBC specimens, can also be genetic test materials.
Collapse
Affiliation(s)
- Kenji Notohara
- Department of Anatomic Pathology, Kurashiki Central Hospital, 1-1-1 Miwa, Kurashiki, 710-8602, Japan.
| | - Kaori Nakamura
- Pathological Laboratory, Division of Medical Technology, Kurashiki Central Hospital, Kurashiki, Japan
| |
Collapse
|
38
|
Urganci N, Kepil N, Ergun S, Bakkaloglu OK. The relationship between DNA mismatch repair gene and other prognostic parameters in pancreatic adenocarcinoma. J Surg Oncol 2024; 129:876-884. [PMID: 38173349 DOI: 10.1002/jso.27579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024]
Abstract
The aim of the study was to determine DNA mismatch repair (MMR) proteins by immunohistochemically using MLH1, MSH2, MSH6, and PMS2 antibodies in patients diagnosed as pancreatic ductal adenocarcinoma and to assess its relationship with histopathological and clinical prognostic parameters. Fifty cases with a diagnosis of pancreatic ductal adenocarcinoma who underwent surgical resection, were included in the study. Demographic and histopathological features of the patients were collected from the medical records. The relationships between microsatellite status and prognostic parameters were determined. The mean age of the patients was 66.5 ± 9.5 years (range: 47-87) and male/female ratio was 1.63 (31/19). No errors were detected in DNA MMR proteins in any of the cases, and were classified as microsatellite stable. The mean tumor diameter was 4.01 ± 1.77 cm and 74% of the tumors were localized in the pancreatic head. All of the cases had lymphatic invasion, whereas vascular invasion was detected in only 78% and perineural invasion in 98% of the patients. When the relationship between prognostic parameters and survival was evaluated, statistically significant correlation was observed in patient age and histopathological parameters such as tumor diameter, status of surgical margins, and vascular invasion (p < 0.05). Age, tumor size, presence of tumor at surgical margins, vascular invasion, and adjuvant treatment were correlated with survival. Although microsatellite instability was not detected in our cases, it is important to determine the microsatellite status by immunohistochemistry for predicting the chemotherapy response and determining the immunotherapy option in pancreatic adenocarcinomas.
Collapse
Affiliation(s)
- Nil Urganci
- Department of Pathology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Nuray Kepil
- Department of Pathology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Sefa Ergun
- Department of General Surgery, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Oguz Kaan Bakkaloglu
- Department of Gastroenterology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
39
|
Matejcic M, Teer JK, Hoehn HJ, Diaz DB, Shankar K, Gong J, Nguyen NT, Lorona N, Coppola D, Fulmer C, Saglam O, Jiang K, Cress D, Muñoz-Antonia T, Flores I, Gordian E, Oliveras Torres JA, Felder SI, Sanchez JA, Fleming J, Siegel EM, Freedman JA, Dutil J, Stern MC, Fridley BL, Figueiredo JC, Schmit SL. Spectrum of somatic mutational features of colorectal tumors in ancestrally diverse populations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.11.24303880. [PMID: 38558992 PMCID: PMC10980113 DOI: 10.1101/2024.03.11.24303880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Ancestrally diverse and admixed populations, including the Hispanic/Latino/a/x/e community, are underrepresented in cancer genetic and genomic studies. Leveraging the Latino Colorectal Cancer Consortium, we analyzed whole exome sequencing data on tumor/normal pairs from 718 individuals with colorectal cancer (128 Latino, 469 non-Latino) to map somatic mutational features by ethnicity and genetic ancestry. Global proportions of African, East Asian, European, and Native American ancestries were estimated using ADMIXTURE. Associations between global genetic ancestry and somatic mutational features across genes were examined using logistic regression. TP53 , APC , and KRAS were the most recurrently mutated genes. Compared to non-Latino individuals, tumors from Latino individuals had fewer KRAS (OR=0.64, 95%CI=0.41-0.97, p=0.037) and PIK3CA mutations (OR=0.55, 95%CI=0.31-0.98, p=0.043). Genetic ancestry was associated with presence of somatic mutations in 39 genes (FDR-adjusted LRT p<0.05). Among these genes, a 10% increase in African ancestry was associated with significantly higher odds of mutation in KNCN (OR=1.34, 95%CI=1.09-1.66, p=5.74×10 -3 ) and TMEM184B (OR=1.53, 95%CI=1.10-2.12, p=0.011). Among RMGs, we found evidence of association between genetic ancestry and mutation status in CDC27 (LRT p=0.0084) and between SMAD2 mutation status and AFR ancestry (OR=1.14, 95%CI=1.00-1.30, p=0.046). Ancestry was not associated with tumor mutational burden. Individuals with above-average Native American ancestry had a lower frequency of microsatellite instable (MSI-H) vs microsatellite stable tumors (OR=0.45, 95%CI=0.21-0.99, p=0.048). Our findings provide new knowledge about the relationship between ancestral haplotypes and somatic mutational profiles that may be useful in developing precision medicine approaches and provide additional insight into genomic contributions to cancer disparities. Significance Our data in ancestrally diverse populations adds essential information to characterize mutational features in the colorectal cancer genome. These results will help enhance equity in the development of precision medicine strategies.
Collapse
|
40
|
Sadozai H, Acharjee A, Kayani HZ, Gruber T, Gorczynski RM, Burke B. High hypoxia status in pancreatic cancer is associated with multiple hallmarks of an immunosuppressive tumor microenvironment. Front Immunol 2024; 15:1360629. [PMID: 38510243 PMCID: PMC10951397 DOI: 10.3389/fimmu.2024.1360629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 02/12/2024] [Indexed: 03/22/2024] Open
Abstract
Introduction Pancreatic ductal adenocarcinoma (PDAC), the most common form of pancreatic cancer, is a particularly lethal disease that is often diagnosed late and is refractory to most forms of treatment. Tumour hypoxia is a key hallmark of PDAC and is purported to contribute to multiple facets of disease progression such as treatment resistance, increased invasiveness, metabolic reprogramming, and immunosuppression. Methods We used the Buffa gene signature as a hypoxia score to profile transcriptomics datasets from PDAC cases. We performed cell-type deconvolution and gene expression profiling approaches to compare the immunological phenotypes of cases with low and high hypoxia scores. We further supported our findings by qPCR analyses in PDAC cell lines cultured in hypoxic conditions. Results First, we demonstrated that this hypoxia score is associated with increased tumour grade and reduced survival suggesting that this score is correlated to disease progression. Subsequently, we compared the immune phenotypes of cases with high versus low hypoxia score expression (HypoxiaHI vs. HypoxiaLOW) to show that high hypoxia is associated with reduced levels of T cells, NK cells and dendritic cells (DC), including the crucial cDC1 subset. Concomitantly, immune-related gene expression profiling revealed that compared to HypoxiaLOW tumours, mRNA levels for multiple immunosuppressive molecules were notably elevated in HypoxiaHI cases. Using a Random Forest machine learning approach for variable selection, we identified LGALS3 (Galectin-3) as the top gene associated with high hypoxia status and confirmed its expression in hypoxic PDAC cell lines. Discussion In summary, we demonstrated novel associations between hypoxia and multiple immunosuppressive mediators in PDAC, highlighting avenues for improving PDAC immunotherapy by targeting these immune molecules in combination with hypoxia-targeted drugs.
Collapse
Affiliation(s)
- Hassan Sadozai
- Centre for Health and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Animesh Acharjee
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Hateem Z. Kayani
- Centre for Health and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Thomas Gruber
- Independent Scholar, National Coalition of Independent Scholars, Visp, Switzerland
| | | | - Bernard Burke
- Centre for Health and Life Sciences, Coventry University, Coventry, United Kingdom
| |
Collapse
|
41
|
Tamaki A, Kato T, Sakurai Y, Sato K, Adachi K, Tadehara M, Kogami T, Matsushita M, Hoshino A, Sanoyama I, Numata Y, Umezawa A, Ichinoe M, Ichihara M, Kusano C, Murakumo Y. REV7 is involved in outcomes of platinum-based chemotherapy in pancreatic cancer by controlling the DNA damage response. Cancer Sci 2024; 115:660-671. [PMID: 38130032 PMCID: PMC10859597 DOI: 10.1111/cas.16044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023] Open
Abstract
REV7 is a multifunctional protein implicated in various biological processes, including DNA damage response. REV7 expression in human cancer cells affects their sensitivity to DNA-damaging agents. In the present study, we investigated the significance of REV7 in pancreatic ductal adenocarcinoma (PDAC). REV7 expression was immunohistochemically examined in 92 resected PDAC specimens and 60 endoscopic ultrasound-guided fine-needle aspiration biopsy (EUS-FNAB) specimens of unresectable PDAC treated with platinum-based chemotherapy, and its association with clinicopathologic features was analyzed. Although REV7 expression was not significantly associated with the progression of primary tumors (T-factor and Stage) in either resected or unresectable PDAC, decreased levels of REV7 expression in EUS-FNAB specimens of unresectable PDAC were significantly associated with better outcomes of platinum-based chemotherapy and a favorable prognosis. REV7-deficient PDAC cell lines showed suppressed cell growth and enhanced sensitivity to cisplatin in vitro. Tumor-bearing mice generated using REV7-deficient PDAC cell lines also showed enhanced sensitivity to cisplatin in vivo. RNA sequencing analysis using WT and REV7-deficient PDAC cell lines revealed that REV7 inactivation promoted the downregulation of genes involved in the DNA repair and the upregulation of genes involved in apoptosis. Our results indicate that decreased expression of REV7 is associated with better outcomes of platinum-based chemotherapy in PDAC by suppressing the DNA damage response. It is also suggested that REV7 is a useful biomarker for predicting the outcome of platinum-based chemotherapy and the prognosis of unresectable PDAC and is a potential target for PDAC treatment.
Collapse
Affiliation(s)
- Akihiro Tamaki
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Takuya Kato
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Yasutaka Sakurai
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Keita Sato
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Kai Adachi
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Masayoshi Tadehara
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Taro Kogami
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Masahiro Matsushita
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Akiyoshi Hoshino
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Itaru Sanoyama
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Yoshiko Numata
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Atsuko Umezawa
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Masaaki Ichinoe
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Masatoshi Ichihara
- Department of Biomedical Sciences, College of Life and Health SciencesChubu UniversityKasugaiJapan
| | - Chika Kusano
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Yoshiki Murakumo
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| |
Collapse
|
42
|
Sakakida T, Ishikawa T, Doi T, Morita R, Kataoka S, Miyake H, Yamaguchi K, Moriguchi M, Sogame Y, Yasuda H, Iwasaku M, Konishi H, Takayama K, Itoh Y. Genomic profile and clinical features of MSI-H and TMB-high pancreatic cancers: real-world data from C-CAT database. J Gastroenterol 2024; 59:145-156. [PMID: 38006445 DOI: 10.1007/s00535-023-02058-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 11/01/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND Microsatellite instability high (MSI-H) and tumor mutational burden high (TMB-high) pancreatic cancer are rare, and information is lacking. Based on the C-CAT database, we analyzed the clinical and genomic characteristics of patients with these subtypes. METHODS We retrospectively reviewed data on 2206 patients with unresectable pancreatic adenocarcinoma enrolled in C-CAT between July 2019 and January 2022. The clinical features, proportion of genomic variants classified as oncogenic/pathogenic in C-CAT, overall response rate (ORR), disease control rate (DCR), and time to treatment failure (TTF) of chemotherapy as first-line treatment were evaluated. RESULTS Numbers of patients with MSI-H and TMB-high were 7 (0.3%) and 39 (1.8%), respectively. All MSI-H patients were TMB-high. MSI-H and TMB-high patients harbored more mismatch repair genes, such as MSH2, homologous recombination-related genes, such as ATR and BRCA2, and other genes including BRAF, KMT2D, and SMARCA4. None of the 6 MSI-H patients who received chemotherapy achieved a clinical response, including 4 patients treated with gemcitabine plus nab-paclitaxel (GnP) therapy, whose DCR was significantly lower than that of microsatellite stable (MSS) patients (0 vs. 67.0%, respectively, p = 0.01). Among the TMB-high and TMB-low groups, no significant differences were shown in ORR, DCR (17.1 vs. 23.1% and 57.1 vs. 63.1%, respectively), or median TTF (25.9 vs. 28.0 weeks, respectively) of overall first-line chemotherapy. CONCLUSIONS MSI-H and TMB-high pancreatic cancers showed some distinct genomic and clinical features from our real-world data. These results suggest the importance of adapting optimal treatment strategies according to the genomic alterations.
Collapse
Affiliation(s)
- Tomoki Sakakida
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Hirokoji agaru, Kawaramachi Street, Kamigyoku, Kyoto, Kyoto, 602-8566, Japan
- Department of Cancer Genome Medical Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Ishikawa
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Hirokoji agaru, Kawaramachi Street, Kamigyoku, Kyoto, Kyoto, 602-8566, Japan.
- Department of Cancer Genome Medical Center, Kyoto Prefectural University of Medicine, Kyoto, Japan.
- Outpatient Oncology Unit, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Toshifumi Doi
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Hirokoji agaru, Kawaramachi Street, Kamigyoku, Kyoto, Kyoto, 602-8566, Japan
- Department of Cancer Genome Medical Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ryuichi Morita
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Hirokoji agaru, Kawaramachi Street, Kamigyoku, Kyoto, Kyoto, 602-8566, Japan
- Department of Cancer Genome Medical Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Seita Kataoka
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Hirokoji agaru, Kawaramachi Street, Kamigyoku, Kyoto, Kyoto, 602-8566, Japan
| | - Hayato Miyake
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Hirokoji agaru, Kawaramachi Street, Kamigyoku, Kyoto, Kyoto, 602-8566, Japan
| | - Kanji Yamaguchi
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Hirokoji agaru, Kawaramachi Street, Kamigyoku, Kyoto, Kyoto, 602-8566, Japan
| | - Michihisa Moriguchi
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Hirokoji agaru, Kawaramachi Street, Kamigyoku, Kyoto, Kyoto, 602-8566, Japan
| | - Yoshio Sogame
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Hirokoji agaru, Kawaramachi Street, Kamigyoku, Kyoto, Kyoto, 602-8566, Japan
| | - Hiroaki Yasuda
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Hirokoji agaru, Kawaramachi Street, Kamigyoku, Kyoto, Kyoto, 602-8566, Japan
| | - Masahiro Iwasaku
- Department of Cancer Genome Medical Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hideyuki Konishi
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Hirokoji agaru, Kawaramachi Street, Kamigyoku, Kyoto, Kyoto, 602-8566, Japan
| | - Koichi Takayama
- Department of Cancer Genome Medical Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Outpatient Oncology Unit, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Hirokoji agaru, Kawaramachi Street, Kamigyoku, Kyoto, Kyoto, 602-8566, Japan
| |
Collapse
|
43
|
Trembath HE, Yeh JJ, Lopez NE. Gastrointestinal Malignancy: Genetic Implications to Clinical Applications. Cancer Treat Res 2024; 192:305-418. [PMID: 39212927 DOI: 10.1007/978-3-031-61238-1_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Advances in molecular genetics have revolutionized our understanding of the pathogenesis, progression, and therapeutic options for treating gastrointestinal (GI) cancers. This chapter provides a comprehensive overview of the molecular landscape of GI cancers, focusing on key genetic alterations implicated in tumorigenesis across various anatomical sites including GIST, colon and rectum, and pancreas. Emphasis is placed on critical oncogenic pathways, such as mutations in tumor suppressor genes, oncogenes, chromosomal instability, microsatellite instability, and epigenetic modifications. The role of molecular biomarkers in predicting prognosis, guiding treatment decisions, and monitoring therapeutic response is discussed, highlighting the integration of genomic profiling into clinical practice. Finally, we address the evolving landscape of precision oncology in GI cancers, considering targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Hannah E Trembath
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA
| | - Jen Jen Yeh
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA
| | - Nicole E Lopez
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA.
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA.
| |
Collapse
|
44
|
Lote H, Chau I. Immunotherapy in Gastrointestinal Cancers. Cancer Treat Res 2024; 192:277-303. [PMID: 39212926 DOI: 10.1007/978-3-031-61238-1_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Immunotherapy has revolutionised cancer treatment over the past decade. Long-term durable responses can be achieved in some cancer patient populations that were previously facing terminal disease. In this chapter, we summarise current phase 3 clinical trial evidence for the use of immunotherapy in gastrointestinal cancers (oesophageal squamous cell carcinoma, oesophago-gastric adenocarcinoma, pancreatic cancer, biliary cancer, hepatocellular carcinoma, colorectal cancer, and squamous cell cancer of the anus). We discuss meaningful biomarkers used in clinical trials to select patients most likely to benefit from immunotherapy, such as mismatch-repair deficiency (MMRd)/microsatellite instability (MSI) and programmed-death-ligand-1 (PD-L1) immunohistochemistry (IHC) expression. Clinical questions are arising regarding the role of immunotherapy in the adjuvant/perioperative setting, optimal timing of surgery in patients who respond to immunotherapy, and toxicities specific to patients with gastrointestinal malignancies. We outline the current landscape and future horizon of immunotherapy in gastrointestinal cancers, such as strategies to increase effectiveness of checkpoint blockade through combinations with other checkpoint inhibitors, cytotoxic chemotherapy, targeted agents, radiotherapy, CAR-T therapy, and cancer vaccines.
Collapse
Affiliation(s)
- Hazel Lote
- The Royal Marsden Hospital NHS Foundation Trust, London and Sutton, UK
- Institute of Cancer Research, Sutton, UK
| | - Ian Chau
- The Royal Marsden Hospital NHS Foundation Trust, London and Sutton, UK.
| |
Collapse
|
45
|
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a rising incidence and is one of the most lethal human malignancies. Much is known regarding the biology and pathophysiology of PDAC, but translating this knowledge to the clinic to improve patient outcomes has been challenging. In this Review, we discuss advances and practice-changing trials for PDAC. We briefly review therapeutic failures as well as ongoing research to refine the standard of care, including novel biomarkers and clinical trial designs. In addition, we highlight contemporary areas of research, including poly(ADP-ribose) polymerase inhibitors, KRAS-targeted therapies and immunotherapies. Finally, we discuss the future of pancreatic cancer research and areas for improvement in the next decade.
Collapse
Affiliation(s)
- Z Ian Hu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eileen M O'Reilly
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
46
|
Pourali G, Donyadideh G, Mehrabadi S, Hamid F, Hassanian SM, Ferns GA, Khazaei M, Avan A. Clinical practice guidelines for interventional treatment of pancreatic cancer. RECENT ADVANCES IN NANOCARRIERS FOR PANCREATIC CANCER THERAPY 2024:345-373. [DOI: 10.1016/b978-0-443-19142-8.00008-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
47
|
Mahadevia H, Uson Junior PLS, Wang J, Borad M, Babiker H. An overview of up-and-coming immune checkpoint inhibitors for pancreatic cancer. Expert Opin Pharmacother 2024; 25:79-90. [PMID: 38193476 DOI: 10.1080/14656566.2024.2304125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 01/08/2024] [Indexed: 01/10/2024]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) targeting programmed cell death protein-1 (PD-1/PD-L1) pathway as well as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) have demonstrated substantial potential in several malignancies. Pancreatic adenocarcinoma (PC) still carries a high mortality despite tremendous advances in the anti-cancer arsenal. AREAS COVERED In this review, we discuss completed and ongoing studies on various ICIs in PC. ICIs have not yielded significant benefits as monotherapy. However, the combination with currently utilized therapies as well as with several other newer forms of therapy has delineated encouraging results. Larger trials are currently underway to definitively characterize the utility of ICIs in the treatment algorithm of PC. ICIs are approved for cancers with mismatch repair deficiency (dMMR) or microsatellite instability-high tumors (MSI-H) as a tumor-agnostic treatment strategy usually referred to as hot tumors. EXPERT OPINION Studies evaluating different drugs to transform the tumor microenvironment (TME) from 'cold' to 'hot' have not shown promise in PC. There still needs to be more prospective trials evaluating the efficacy of the combination of ICIs with different therapeutic modalities in PC that can augment the immunogenic potential of those 'cold' tumors. Exploratory biomarker analysis may help us identify those subsets of PC patients who may particularly benefit from ICIs.
Collapse
Affiliation(s)
- Himil Mahadevia
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, MO, USA
- Department of Internal Medicine, Division of Hematology-Oncology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Pedro Luiz Serrano Uson Junior
- Department of Internal Medicine, Division of Hematology-Oncology, Hospital Israelita Albert Einstein, Sao Paulo, SP, Brazil
- Department of Internal Medicine, Division of Hematology-Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Jing Wang
- Department of Internal Medicine, Division of Hematology-Oncology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Mitesh Borad
- Department of Internal Medicine, Division of Hematology-Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Hani Babiker
- Department of Internal Medicine, Division of Hematology-Oncology, Mayo Clinic Florida, Jacksonville, FL, USA
| |
Collapse
|
48
|
Matsuoka T, Yashiro M. Current status and perspectives of genetic testing in gastrointestinal cancer (Review). Oncol Lett 2024; 27:21. [PMID: 38058469 PMCID: PMC10696628 DOI: 10.3892/ol.2023.14155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/30/2023] [Indexed: 12/08/2023] Open
Abstract
Genetic testing has become widespread in daily medical care for gastrointestinal (GI) cancers. However, unlike breast cancer and non-small cell lung cancer, in which personalized medicine targeting various driver genes is standardized, the incidence of targeted gene abnormalities in GI cancers is low. Nevertheless, such abnormalities may be linked to therapeutic agents and the further development of therapeutic agents for personalized medicine for GI cancers is desired. A liquid biopsy is of great benefit in offering clinical decision support, in applications such as GI cancer screening, surgical interventions, monitoring disease status and enhancing patient survival outcomes, all of which would contribute to personalized medicine. Germline genetic testing is required for several types of GI cancer, which shows clinical indications of hereditary predisposition. The increasing use of multigene panel testing has redefined gene-cancer associations, and consequently the estimate of cancer risk that vary from low to high penetrance. Comprehensive genetic testing can enable the detection of novel treatment targets and the discovery of undefined multiple diagnostic/predictive markers, which may enhance the molecular-level understanding of GI cancers. Genetic testing can also aid the design of more appropriate and adequate genomic-driven therapies for patients who may benefit from other standardized therapeutic methods.
Collapse
Affiliation(s)
- Tasuku Matsuoka
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 5458585, Japan
| | - Masakazu Yashiro
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 5458585, Japan
- Institute of Medical Genetics, Osaka Metropolitan University, Osaka 5458585, Japan
| |
Collapse
|
49
|
Behrens D, Pfohl U, Conrad T, Becker M, Brzezicha B, Büttner B, Wagner S, Hallas C, Lawlor R, Khazak V, Linnebacher M, Wartmann T, Fichtner I, Hoffmann J, Dahlmann M, Walther W. Establishment and Thorough Characterization of Xenograft (PDX) Models Derived from Patients with Pancreatic Cancer for Molecular Analyses and Chemosensitivity Testing. Cancers (Basel) 2023; 15:5753. [PMID: 38136299 PMCID: PMC10741928 DOI: 10.3390/cancers15245753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Patient-derived xenograft (PDX) tumor models are essential for identifying new biomarkers, signaling pathways and novel targets, to better define key factors of therapy response and resistance mechanisms. Therefore, this study aimed at establishing pancreas carcinoma (PC) PDX models with thorough molecular characterization, and the identification of signatures defining responsiveness toward drug treatment. In total, 45 PC-PDXs were generated from 120 patient tumor specimens and the identity of PDX and corresponding patient tumors was validated. The majority of engrafted PDX models represent ductal adenocarcinomas (PDAC). The PDX growth characteristics were assessed, with great variations in doubling times (4 to 32 days). The mutational analyses revealed an individual mutational profile of the PDXs, predominantly showing alterations in the genes encoding KRAS, TP53, FAT1, KMT2D, MUC4, RNF213, ATR, MUC16, GNAS, RANBP2 and CDKN2A. Sensitivity of PDX toward standard of care (SoC) drugs gemcitabine, 5-fluorouracil, oxaliplatin and abraxane, and combinations thereof, revealed PDX models with sensitivity and resistance toward these treatments. We performed correlation analyses of drug sensitivity of these PDX models and their molecular profile to identify signatures for response and resistance. This study strongly supports the importance and value of PDX models for improvement in therapies of PC.
Collapse
Affiliation(s)
- Diana Behrens
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Ulrike Pfohl
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Theresia Conrad
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Michael Becker
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Bernadette Brzezicha
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Britta Büttner
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Silvia Wagner
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Cora Hallas
- Institut für Hämatopathologie, Fangdieckstr. 75, 22547 Hamburg, Germany
| | - Rita Lawlor
- ARC-Net Research Center, University and Hospital Trust of Verona, Piazzale A. Scuro 10, 37134 Verona, Italy
| | | | - Michael Linnebacher
- Clinic of General Surgery, Molecular Oncology and Immunotherapy, University Medical Center Rostock, 18057 Rostock, Germany
| | - Thomas Wartmann
- University Clinic for General, Visceral, Vascular and Transplantation Surgery, Faculty of Medicine, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Iduna Fichtner
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Jens Hoffmann
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Mathias Dahlmann
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Wolfgang Walther
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
- Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin, Lindenberger Weg 80, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| |
Collapse
|
50
|
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. Pancreatic cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2023; 34:987-1002. [PMID: 37678671 DOI: 10.1016/j.annonc.2023.08.009] [Citation(s) in RCA: 173] [Impact Index Per Article: 86.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Affiliation(s)
- T Conroy
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Vandoeuvre-lès-Nancy; APEMAC, équipe MICS, Université de Lorraine, Nancy, France
| | - P Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - V Vilgrain
- Centre de Recherche sur l'Inflammation U 1149, Université Paris Cité, Paris; Department of Radiology, Beaujon Hospital, APHP Nord, Clichy, France
| | - A Lamarca
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - T Seufferlein
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - E M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - T Hackert
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - T Golan
- Gastrointestinal Unit, Oncology Institute, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - G Prager
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - K Haustermans
- Department of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - A Vogel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - M Ducreux
- Université Paris-Saclay, Gustave Roussy, Inserm Unité Dynamique des Cellules Tumorales, Villejuif, France
| |
Collapse
|