1
|
Li Y, Xiong X, Liu X, Xu M, Yang B, Li X, Li Y, Lin B, Xu B. Predicting BRCA mutation and stratifying targeted therapy response using multimodal learning: a multicenter study. Ann Med 2024; 56:2399759. [PMID: 39258876 PMCID: PMC11391871 DOI: 10.1080/07853890.2024.2399759] [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: 03/14/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND The status of BRCA1/2 genes plays a crucial role in the treatment decision-making process for multiple cancer types. However, due to high costs and limited resources, a demand for BRCA1/2 genetic testing among patients is currently unmet. Notably, not all patients with BRCA1/2 mutations achieve favorable outcomes with poly (ADP-ribose) polymerase inhibitors (PARPi), indicating the necessity for risk stratification. In this study, we aimed to develop and validate a multimodal model for predicting BRCA1/2 gene status and prognosis with PARPi treatment. METHODS We included 1695 slides from 1417 patients with ovarian, breast, prostate, and pancreatic cancers across three independent cohorts. Using a self-attention mechanism, we constructed a multi-instance attention model (MIAM) to detect BRCA1/2 gene status from hematoxylin and eosin (H&E) pathological images. We further combined tissue features from the MIAM model, cell features, and clinical factors (the MIAM-C model) to predict BRCA1/2 mutations and progression-free survival (PFS) with PARPi therapy. Model performance was evaluated using area under the curve (AUC) and Kaplan-Meier analysis. Morphological features contributing to MIAM-C were analyzed for interpretability. RESULTS Across the four cancer types, MIAM-C outperformed the deep learning-based MIAM in identifying the BRCA1/2 genotype. Interpretability analysis revealed that high-attention regions included high-grade tumors and lymphocytic infiltration, which correlated with BRCA1/2 mutations. Notably, high lymphocyte ratios appeared characteristic of BRCA1/2 mutations. Furthermore, MIAM-C predicted PARPi therapy response (log-rank p < 0.05) and served as an independent prognostic factor for patients with BRCA1/2-mutant ovarian cancer (p < 0.05, hazard ratio:0.4, 95% confidence interval: 0.16-0.99). CONCLUSIONS The MIAM-C model accurately detected BRCA1/2 gene status and effectively stratified prognosis for patients with BRCA1/2 mutations.
Collapse
Affiliation(s)
- Yi Li
- School of Medicine, Chongqing University, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaomin Xiong
- School of Medicine, Chongqing University, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaohua Liu
- Bioengineering College of Chongqing University, Chongqing, China
| | - Mengke Xu
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
| | - Boping Yang
- Department of General Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Xiaoju Li
- Department of Pathology, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Yu Li
- Department of Pathology, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Bo Lin
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
| | - Bo Xu
- School of Medicine, Chongqing University, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
2
|
Feng X, Chai YH, Jiang KX, Jiang WB, Chen WC, Pan Y. Bibliometric analysis of olaparib and pancreatic cancer from 2009 to 2022: A global perspective. World J Gastrointest Oncol 2024; 16:4489-4505. [DOI: 10.4251/wjgo.v16.i11.4489] [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: 06/04/2024] [Revised: 08/24/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Genetic screening for breast cancer gene 1 (BRCA)1/2 mutations can inform breast/ovarian/pancreatic cancer patients of suitable therapeutic interventions. Four to seven percent of pancreatic cancer patients have germline BRCA mutations. BRCA genes aid in DNA repair, especially homologous recombination, which impacts genomic stability and cancer cell growth. BRCA1 regulates the cell cycle, ubiquitination, and chromatin remodeling, whereas BRCA2 stimulates the immune response. They predict the efficacy of platinum chemotherapy or polymerase (PARP) inhibitors such as olaparib.
AIM To determine the trends and future directions in the use of olaparib for pancreatic cancer treatment.
METHODS To evaluate the trends in how olaparib works in pancreatic cancer, we performed a bibliometric analysis. One hundred and ninety-six related publications were accessed from the Web of Science Core Collection and were published between 2009 and 2022. The analytic parameters included publications, related citations, productive countries and institutes, influential authors, and keyword development.
RESULTS This study visualizes and discusses the current research, including the present global trends and future directions in olaparib and pancreatic cancer. Overall, this study sheds light on optimizing the use of olaparib in pancreatic cancer treatment, offering valuable guidance for researchers in this field.
CONCLUSION Our findings identified trends in olaparib and pancreatic cancer, with China and the USA leading and with global cooperation tightening. O'Reilly EM's team and Memorial Sloan-Kettering had the highest output. The Journal of Clinical Oncology was the most cited journal. More PARP inhibitors are emerging, and combination therapy is suggested for future therapeutic trends.
Collapse
Affiliation(s)
- Xu Feng
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Yi-Han Chai
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Ke-Xin Jiang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Wen-Bin Jiang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Wen-Chao Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Yu Pan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| |
Collapse
|
3
|
McDonald HG, Kennedy A, Solomon AL, Williams CM, Reagan AM, Cassim E, Harper M, Burke E, Armstrong T, Gosky M, Cavnar M, Pandalai PK, Barry-Hundeyin M, Patel R, Nutalapati S, Moss J, Hull PC, Kolesar J, Pickarski JC, Kim J. Development of a Novel Protocol for Germline Testing in Pancreatic Cancer. Ann Surg Oncol 2024; 31:7705-7712. [PMID: 39133448 DOI: 10.1245/s10434-024-16011-3] [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: 05/20/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Guidelines now recommend universal germline genetic testing (GGT) for all pancreatic ductal adenocarcinoma (PDAC) patients. Testing provides information on actionable pathogenic variants and guides management of patients and family. Since traditional genetic counseling (GC) models are time-intensive and GC resources are sparse, new approaches are needed to comply with guidelines without overwhelming available resources. METHODS A novel protocol was developed for physician-led GGT. Completed test kits were delivered to the GC team, who maintained a prospective database and mailed all orders. If results revealed pathogenic variants for PDAC, patients were offered comprehensive GC, whereas negative and variant of uncertain significance (VUS) test results were reported to patients via brief calls. RESULTS During protocol implementation between January 2020 and December 2022, 310 (81.5%) patients underwent GGT, with a physician compliance rate of 82.6% and patient compliance rate of 98.7%. Of 310 patients tested, 44 (14.2%) patients had detection of pathogenic variants, while 83 (26.8%) patients had VUS. Pathogenic variants included BRCA1/BRCA2/PALB2 (n = 18, 5.8%), ATM (n = 9, 2.9%), CFTR (n = 4, 1.3%), EPCAM/MLH1/MSH2/MSH6/PMS2 (n = 3, 1.0%), and CDKN2A (n = 2, 0.7%). The GC team successfully contacted all patients with pathogenic variants to discuss results and offer comprehensive GC. CONCLUSION Our novel protocol facilitated GGT with excellent compliance despite limited GC resources. This framework for GGT allocates GC resources to those patients who would benefit most from GC. As we continue to expand the program, we seek to implement methods to ensure compliance with cascade testing of high-risk family members.
Collapse
Affiliation(s)
- Hannah G McDonald
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Andrew Kennedy
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Angelica L Solomon
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Chelsey M Williams
- Division of Hematology Oncology, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Anna M Reagan
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Emily Cassim
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Megan Harper
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Erin Burke
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Terra Armstrong
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Michael Gosky
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Michael Cavnar
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Prakash K Pandalai
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Mautin Barry-Hundeyin
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Reema Patel
- Division of Hematology Oncology, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Snigdha Nutalapati
- Division of Hematology Oncology, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Jessica Moss
- Division of Hematology Oncology, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Pamela C Hull
- Department of Behavioral Science, University of Kentucky, Lexington, KY, USA
| | - Jill Kolesar
- College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | | | - Joseph Kim
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
4
|
M'Baloula J, Tougeron D, Boilève A, Jeanbert E, Guimbaud R, Ben Abdelghani M, Durand A, Turpin A, Quesada S, Blanc JF, Artru P, Toullec C, Trouilloud I, Pellat A, Touchefeu Y, Pinot J, Caroli-Bosc FX, Taïeb J, Doat S, Bouché O, Védie AL, de Mestier L, Muller M. Olaparib as maintenance therapy in non resectable pancreatic adenocarcinoma associated with homologous recombination deficiency profile: A French retrospective multicentric AGEO real-world study. Eur J Cancer 2024; 212:115051. [PMID: 39366210 DOI: 10.1016/j.ejca.2024.115051] [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: 07/19/2024] [Revised: 09/13/2024] [Accepted: 09/22/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis. The POLO trial showed that olaparib (PARP inhibitor) improved progression-free survival (PFS) but not overall survival (OS), when used as maintenance therapy after ≥ 16 weeks of disease control with first-line platinum-based chemotherapy in patients with germline (g) BRCA 1 or 2 pathogenic variants (PV) metastatic PDAC. However, real-world data on the effectiveness of olaparib are missing. METHODS Patients with unresectable PDAC associated with somatic (s) or (g)BRCA1/2 and (g)non-BRCA-HRD PV (i.e. other homologous recombination deficiency/HRD genes) who were treated with olaparib between 2020-2023 were included. The primary objective was to describe treatment patterns. Secondary exploratory objectives included OS and PFS in patients treated with olaparib according to the POLO trial or not, OS and PFS in patients with (g)HRD PV-associated PDAC versus (s)PVs, olaparib safety profile and factors associated with olaparib poor outcomes. RESULTS Among 85 patients, 45.9 % received olaparib as defined by the POLO trial. No difference in OS and PFS was observed between patients who received olaparib according to the POLO trial versus not. Patients with (g)HRD PV-associated PDAC had better OS compared to others (22.3 versus 10.5 months, p = 0.038). Factors associated with olaparib poor outcomes included a high neutrophil-to-lymphocyte ratio and the use of olaparib outside the recommendations of the POLO trial. Few grade ≥ 3 adverse events were reported (9.4 %). CONCLUSION Patients with (g)HRD PV-associated PDAC had longer OS than those with (s)HRD PV. Olaparib use beyond the scope of the POLO trial was associated with poor outcomes.
Collapse
Affiliation(s)
- Jeannie M'Baloula
- University of Lorraine, Department of Hepato-Gastroenterology and digestive oncology, Nancy University Hospital, Nancy, France
| | - David Tougeron
- Department of Hepatology and Gastroenterology, Poitiers University Hospital, Poitiers, France
| | - Alice Boilève
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Elodie Jeanbert
- DRCI, MPI Department, Methodology, Data Management and Statistics Unit, Nancy University Hospital, Nancy, France
| | - Rosine Guimbaud
- Digestive Medical Oncology, Toulouse University Hospital, Toulouse, France
| | | | - Alice Durand
- Department of Gastroenterology and digestive oncology, Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon, France
| | - Anthony Turpin
- Medical Oncology Department, Lille University Hospital, University of Lille, Lille, France
| | - Stanislas Quesada
- Medical Oncology Department, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Jean Frédéric Blanc
- Department of Gastroenterology and digestive oncology, Hôpital Haut-Lévêque, Bordeaux University Hospital, Bordeaux, France
| | - Pascal Artru
- kHepato-gastroenterology department, Hôpital Jean-Mermoz, Lyon, France
| | - Clémence Toullec
- Medical Oncology Department, Sainte Catherine Institut du Cancer Avignon-Provence, Avignon, France
| | - Isabelle Trouilloud
- Department of Medical Oncology, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, Sorbonne University, Paris, France
| | - Anna Pellat
- Department of Gastroenterology, endoscopy and digestive oncology, Assistance Publique-Hôpitaux de Paris, Cochin University Hospital, Paris, France
| | - Yann Touchefeu
- Hepato-gastroenterology Department, Nantes University Hospital, Nantes, Institut des Maladies de l'Appareil Digestif (IMAD), Inserm CIC, 1413 Nantes, France
| | - Julien Pinot
- Department of Medical Oncology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | | | - Julien Taïeb
- Department of Gastroenterology and digestive oncology, Assistance Publique-Hôpitaux de Paris, Georges Pompidou European Hospital, CARPEM comprehensive cancer center, Université Paris Cité, Paris, France
| | - Solène Doat
- Department of Gastroenterology, Assistance Publique-Hôpitaux de Paris, Pitié-Salpetrière Hospital, Paris, France
| | - Olivier Bouché
- Department of Hepato-Gastroenterology and digestive oncology, Robert-Debré University Hospital, Reims, France
| | - Anne Laure Védie
- University Paris-Cité, Department of Pancreatology and digestive oncology, Beaujon Hospital (AP-HP), Clichy, France
| | - Louis de Mestier
- University Paris-Cité, Department of Pancreatology and digestive oncology, Beaujon Hospital (AP-HP), Clichy, France
| | - Marie Muller
- University of Lorraine, Department of Hepato-Gastroenterology and digestive oncology, Nancy University Hospital, Nancy, France.
| |
Collapse
|
5
|
Miao R, Blue K, Sommerer K, Shah A, Bottiglieri S, del Cueto A, Berry DK, Ho TT, Hicks JK, Kim DW. PARP Inhibitors in Pancreatic Cancer with Homologous Recombination Repair Gene Mutations: A Single-Institution Experience. Cancers (Basel) 2024; 16:3447. [PMID: 39456541 PMCID: PMC11505755 DOI: 10.3390/cancers16203447] [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: 09/01/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Limited data are available regarding the anticancer activity of PARP inhibitors (PARPis) in pancreatic cancer with mutations in HRR genes other than BRCA and PALB2. METHODS We retrospectively reviewed the clinical characteristics and outcomes of 48 patients with advanced pancreatic cancer harboring pathogenic germline and/or somatic HRR mutations who were treated with PARPis. RESULTS Thirty patients had germline (g)HRR mutations only, twelve had somatic (s)HRR mutations only, and six had concomitant gHRR and sHRR mutations. The objective response rate (ORR) was 22%. The median progression-free survival (mPFS) and overall survival (mOS) were 6.9 and 11.5 months, respectively. Five patients received olaparib in the front-line setting due to borderline performance status. Their ORR was 20%, and their mPFS and mOS were both 11.3 months. The ORR was higher in patients with BRCA or PALB2 mutations (germline or somatic) than in those with non-BRCA/PALB2 mutations. Patients with somatic non-BRCA/PALB2 variants had a shorter mPFS. Patients with concomitant gHRR/sHRR mutations or gHRR mutations alone had a significantly longer mPFS than those with sHRR mutations only. CONCLUSIONS PARP inhibitors may be considered for patients with advanced pancreatic cancer harboring pathogenic alterations of BRCA who cannot tolerate standard chemotherapy. Maintenance PARPis can be considered in selected patients with non-BRCA/non-PALB2 HRR mutations.
Collapse
Affiliation(s)
- Ruoyu Miao
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Kirsten Blue
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Katelyn Sommerer
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Anand Shah
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Sal Bottiglieri
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Alex del Cueto
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (A.d.C.); (D.K.B.); (T.T.H.); (J.K.H.)
| | - Darcy K. Berry
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (A.d.C.); (D.K.B.); (T.T.H.); (J.K.H.)
| | - Teresa T. Ho
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (A.d.C.); (D.K.B.); (T.T.H.); (J.K.H.)
| | - James Kevin Hicks
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (A.d.C.); (D.K.B.); (T.T.H.); (J.K.H.)
| | - Dae Won Kim
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| |
Collapse
|
6
|
Wang J, Yang J, Narang A, He J, Wolfgang C, Li K, Zheng L. Consensus, debate, and prospective on pancreatic cancer treatments. J Hematol Oncol 2024; 17:92. [PMID: 39390609 PMCID: PMC11468220 DOI: 10.1186/s13045-024-01613-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: 08/16/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Pancreatic cancer remains one of the most aggressive solid tumors. As a systemic disease, despite the improvement of multi-modality treatment strategies, the prognosis of pancreatic cancer was not improved dramatically. For resectable or borderline resectable patients, the surgical strategy centered on improving R0 resection rate is consensus; however, the role of neoadjuvant therapy in resectable patients and the optimal neoadjuvant therapy of chemotherapy with or without radiotherapy in borderline resectable patients were debated. Postoperative adjuvant chemotherapy of gemcitabine/capecitabine or mFOLFIRINOX is recommended regardless of the margin status. Chemotherapy as the first-line treatment strategy for advanced or metastatic patients included FOLFIRINOX, gemcitabine/nab-paclitaxel, or NALIRIFOX regimens whereas 5-FU plus liposomal irinotecan was the only standard of care second-line therapy. Immunotherapy is an innovative therapy although anti-PD-1 antibody is currently the only agent approved by for MSI-H, dMMR, or TMB-high solid tumors, which represent a very small subset of pancreatic cancers. Combination strategies to increase the immunogenicity and to overcome the immunosuppressive tumor microenvironment may sensitize pancreatic cancer to immunotherapy. Targeted therapies represented by PARP and KRAS inhibitors are also under investigation, showing benefits in improving progression-free survival and objective response rate. This review discusses the current treatment modalities and highlights innovative therapies for pancreatic cancer.
Collapse
Affiliation(s)
- Junke Wang
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jie Yang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Amol Narang
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jin He
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Christopher Wolfgang
- Department of Surgery, New York University School of Medicine and NYU-Langone Medical Center, New York, NY, USA
| | - Keyu Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA.
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA.
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Multidisciplinary Gastrointestinal Cancer Laboratories Program, the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
7
|
Nicolle R, Canivet C, Palazzo L, Napoléon B, Ayadi M, Pignolet C, Cros J, Gourgou S, Selves J, Torrisani J, Dusetti N, Cordelier P, Buscail L, Bournet B. Predictive genomic and transcriptomic analysis on endoscopic ultrasound-guided fine needle aspiration materials from primary pancreatic adenocarcinoma: a prospective multicentre study. EBioMedicine 2024; 109:105373. [PMID: 39383608 PMCID: PMC11497430 DOI: 10.1016/j.ebiom.2024.105373] [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: 04/18/2024] [Revised: 08/24/2024] [Accepted: 09/17/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND We apply endoscopic ultrasound-guided fine needle aspiration biopsy to cytopathologically diagnose and sample nucleic acids from primary tumours regardless of the disease stage. METHODS 397 patients with proven pancreatic adenocarcinoma were included and followed up in a multicentre prospective study. DNA and mRNA were extracted from materials of primary tumours obtained by endoscopic ultrasound-guided fine needle aspiration biopsy and analysed using targeted deep sequencing and RNAseq respectively. FINDINGS The variant allele frequency of the KRAS mutation was used to evaluate the tumour cellularity, ranging from 15 to 20% in all cells, regardless of the tumour stage. The molecular profile of metastatic primary tumours significantly differed from other types of tumours, more frequently having TP53 mutations (p = 0.0002), less frequently having RNF43 mutations, and possessing more basal-like mRNA component (p = 0.001). Molecular markers associated with improved overall survival were: mutations in homologous recombination deficiency genes in patients who received first-line platinum-based chemotherapy (p = 0.025) and wild-type TP53 gene in patients with locally advanced tumours who received radio-chemotherapy (p = 0.01). The GemPred transcriptomic profile was associated with a significantly better overall survival in patients with locally advanced or metastatic pancreatic cancer who received a gemcitabine-based first-line treatment (p = 0.019). INTERPRETATION The combination of genomic and transcriptomic analyses of primary pancreatic tumours enables us to distinguish metastatic tumours from other tumour types. Our molecular strategy may assist in predicting overall survival outcomes for platinum or gemcitabine-based chemotherapies, as well as radio-chemotherapy. FUNDING Institut National Du Cancer (BCB INCa_7294), CHU of Toulouse, Inserm and Ligue Nationale Contre le Cancer (CIT program).
Collapse
Affiliation(s)
- Rémy Nicolle
- Université Paris Cité, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, Paris F-75018, France
| | - Cindy Canivet
- Service de Gastroentérologie et Pancréatologie, Centre Hospitalier Universitaire de Toulouse-Rangueil (CHU), Toulouse, France
| | | | - Bertrand Napoléon
- Service de Gastroentérologie, Hôpital Privé Jean Mermoz, Ramsay Générale de Santé, Lyon, France
| | - Mira Ayadi
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Camille Pignolet
- Université Paris Cité, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, Paris F-75018, France
| | - Jérôme Cros
- Université Paris Cité, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, Paris F-75018, France; Université Paris Cité, Service d'Anatomopathologie, Centre Hospitalier Universitaire Beaujon/Bichat (APHP), Clichy/Paris, France
| | - Sophie Gourgou
- Institut du Cancer de Montpellier-Val d'Aurelle, Université de Montpellier, Montpellier, France
| | - Janick Selves
- Service d'Anatomopathologie, Institut Universitaire du Cancer-Oncopole de Toulouse, Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Jérôme Torrisani
- Centre de Recherches en Cancérologie de Toulouse, Inserm U1037, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
| | - Nelson Dusetti
- Centre de Recherche sur le Cancer de Marseille, Inserm, CNRS, Institut Paoli-Calmettes, Université Aix-Marseille, Marseille, France
| | - Pierre Cordelier
- Centre de Recherches en Cancérologie de Toulouse, Inserm U1037, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
| | - Louis Buscail
- Service de Gastroentérologie et Pancréatologie, Centre Hospitalier Universitaire de Toulouse-Rangueil (CHU), Toulouse, France; Centre de Recherches en Cancérologie de Toulouse, Inserm U1037, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France.
| | - Barbara Bournet
- Service de Gastroentérologie et Pancréatologie, Centre Hospitalier Universitaire de Toulouse-Rangueil (CHU), Toulouse, France; Centre de Recherches en Cancérologie de Toulouse, Inserm U1037, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
| |
Collapse
|
8
|
Lee JJ, Yeh JJ. Updates in Molecular Profiling of Pancreatic Ductal Adenocarcinoma. Surg Clin North Am 2024; 104:939-950. [PMID: 39237169 PMCID: PMC11377860 DOI: 10.1016/j.suc.2024.04.001] [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] [Indexed: 09/07/2024]
Abstract
Outcomes from pancreatic ductal adenocarcinoma (PDAC) remain poor and better methods of prognostication and therapeutic approaches are needed. Recent advances in cancer genomics have led to the development of molecular subtypes of PDAC associated with clinical outcomes. Current evidence also suggests that the subtypes have differential response to first-line chemotherapy regimens. PDAC is also characterized by different stroma and immune environments. Further work is needed to confirm the utility of these subtypes to predicting response to different systemic therapies.
Collapse
Affiliation(s)
- Jaewon James Lee
- Department of Surgery, University of North Carolina at Chapel Hill, 170 Manning Dr, CB7213, Chapel Hill, NC 27599-7213, USA
| | - Jen Jen Yeh
- Department of Surgery, University of North Carolina at Chapel Hill, 170 Manning Dr, CB7213, Chapel Hill, NC 27599-7213, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 450 West Drive, CB7295, Chapel Hill, NC 27599-7295, USA.
| |
Collapse
|
9
|
Mitri Z, Goodyear SM, Mills G. Strategies for the prevention or reversal of PARP inhibitor resistance. Expert Rev Anticancer Ther 2024; 24:959-975. [PMID: 39145413 DOI: 10.1080/14737140.2024.2393251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 08/16/2024]
Abstract
INTRODUCTION Advances in our understanding of tumor biology shed light on hallmarks of cancer development and progression that include dysregulated DNA damage repair (DDR) machinery. Leveraging the underlying tumor genomic instability and tumor-specific defects in DDR, Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) induced DNA damage emerges as a novel non-chemotherapy therapeutic opportunity. PARPis are currently approved in multiple tumor types, with the largest benefit seen in tumors with homologous recombination repair (HRR) deficiency, including germline and somatic mutations in BRCA1/2 genes (BRCA) and other pathway members such as PALB2 and Rad51c. AREAS COVERED This review article summarizes the current approval landscape and known and proposed mechanisms of resistance to PARPi. Further, therapeutic strategies to overcome PARPi resistance are discussed, including ongoing clinical trials. EXPERT OPINION PARPi have proven to be a safe and effective therapy and represents a cornerstone treatment across multiple solid tumor types. Elucidating innate and acquired mechanisms of resistance, coupled with the emergence of novel therapeutic options to capitalize on the activity of PARPi and prevent or reverse the acquisition of resistance, provides an opportunity to further expand the role of PARPi in cancer therapy.
Collapse
Affiliation(s)
- Zahi Mitri
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Shaun M Goodyear
- Developmental and Cancer Biology, Knight Cancer Institute, Portland, OR, USA
| | - Gordon Mills
- Developmental and Cancer Biology, Knight Cancer Institute, Portland, OR, USA
| |
Collapse
|
10
|
Hewitt DB, Wolfgang CL. The Role of Surgery in "Oligometastatic" Pancreas Cancer. Surg Clin North Am 2024; 104:1065-1081. [PMID: 39237164 DOI: 10.1016/j.suc.2024.04.009] [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: 09/07/2024]
Abstract
The majority of patients diagnosed with pancreatic cancer already have metastatic disease at the time of presentation, which results in a 5-year survival rate of only 13%. However, multiagent chemotherapy regimens can stabilize the disease in select patients with limited metastatic disease. For such patients, a combination of curative-intent therapy and systemic therapy may potentially enhance outcomes compared to using systemic therapy alone. Of note, the evidence supporting this approach is primarily derived from retrospective studies and may carry a significant selection bias. Looking ahead, ongoing prospective trials are exploring the efficacy of curative-intent therapy in managing oligometastatic pancreatic cancer and the implementation of treatment strategies based on specific biomarkers. The emergence of these trials, coupled with the development of less invasive therapeutic modalities, provides hope for patients with oligometastatic pancreatic cancer.
Collapse
Affiliation(s)
- D Brock Hewitt
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The NYU Grossman School of Medicine, 577 1st Avenue, 2nd Floor, New York, NY 10016, USA.
| | - Christopher L Wolfgang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The NYU Grossman School of Medicine, 577 1st Avenue, 2nd Floor, New York, NY 10016, USA
| |
Collapse
|
11
|
Li B, Zhang Q, Castaneda C, Cook S. Targeted Therapies in Pancreatic Cancer: A New Era of Precision Medicine. Biomedicines 2024; 12:2175. [PMID: 39457488 PMCID: PMC11505516 DOI: 10.3390/biomedicines12102175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/15/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a leading cause of cancer mortality in the United States, presents significant treatment challenges due to its late diagnosis and poor prognosis. Despite advances, the five-year survival rates remain dismally low, with only a fraction of patients eligible for potentially curative surgical interventions. This review aims to comprehensively examine the current landscape of targeted therapies in PDAC, focusing on recent developments in precision medicine approaches. We explore various molecular targets, including KRAS mutations, DNA damage repair deficiencies, mismatch repair pathway alterations, and rare genetic fusions. The review discusses emerging therapies, such as PARP inhibitors, immune checkpoint inhibitors, and novel targeted agents, like RET and NTRK inhibitors. We analyze the results of key clinical trials and highlight the potential of these targeted approaches in specific patient subgroups. Recent developments in PDAC research have emphasized precision oncology, facilitated by next-generation sequencing and the identification of genetic and epigenetic alterations. This approach tailors treatments to individual genetic profiles, improving outcomes and reducing side effects. Significant strides have been made in classifying PDAC into various subtypes, enhancing therapeutic precision. The identification of specific mutations in genes like KRAS, along with advancements in targeted therapies, including small molecule inhibitors, offers new hope. Furthermore, emerging therapies targeting DNA repair pathways and immunotherapeutic strategies also show promising results. As research evolves, integrating these targeted therapies with conventional treatments might improve survival rates and quality of life for PDAC patients, underscoring the shift towards a more personalized treatment paradigm.
Collapse
Affiliation(s)
- Bingyu Li
- University of Wisconsin Hospitals and Clinics, Madison, WI 53792-2460, USA; (B.L.); (Q.Z.); (C.C.)
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Qiong Zhang
- University of Wisconsin Hospitals and Clinics, Madison, WI 53792-2460, USA; (B.L.); (Q.Z.); (C.C.)
| | - Claire Castaneda
- University of Wisconsin Hospitals and Clinics, Madison, WI 53792-2460, USA; (B.L.); (Q.Z.); (C.C.)
| | - Shelly Cook
- University of Wisconsin Hospitals and Clinics, Madison, WI 53792-2460, USA; (B.L.); (Q.Z.); (C.C.)
| |
Collapse
|
12
|
Morimoto Y, Takada K, Nakano A, Takeuchi O, Watanabe K, Hirohara M, Masuda Y. Combination of S-1 and the oral ATR inhibitor ceralasertib is effective against pancreatic cancer cells. Cancer Chemother Pharmacol 2024:10.1007/s00280-024-04716-x. [PMID: 39271497 DOI: 10.1007/s00280-024-04716-x] [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: 03/05/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
PURPOSE In our previous study, we found that the Chk1 inhibitor prexasertib enhances the antitumour effect of the oral anticancer drug S-1 against pancreatic cancer cells. In this study, we investigated the effect of combining S-1 and ceralasertib, an oral inhibitor of ATR, which is located upstream of Chk1. Ceralasertib is currently being investigated in multiple clinical trials for various cancers. METHODS The cell-proliferation inhibitory effect was measured by MTT assay, using the pancreatic cancer cell lines BxPC-3, SUIT-2, PANC-1, and MIA PaCa-2, while apoptosis was measured by flow cytometry using PI/Annexin staining. The mechanism underlying the combined effect was analysed using western blotting, and the antitumor effect was analysed using a mouse xenograft model. RESULTS MTT assay revealed that the combination of S-1 and ceralasertib had a synergistic effect, leading to the suppression of cell proliferation. Measurement with PI/Annexin staining revealed that the combination of S-1 and ceralasertib induced apoptosis more efficiently than either drug alone. Western blotting results showed that ceralasertib inhibited S-1-induced activation of ATR and Chk1. The average estimated tumour volume after 3 weeks of administration was 601 mm3 in the S-1 group, 580 mm3 in the ceralasertib group, and 298 mm3 in the combination group. CONCLUSION The combination of S-1 and ceralasertib demonstrated a high antiproliferative effect in inhibiting tumour growth in vitro.
Collapse
Affiliation(s)
- Yoshihito Morimoto
- Center for Education and Research on Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan.
| | - Kimihiko Takada
- Center for Education and Research on Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Ami Nakano
- Center for Education and Research on Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Osamu Takeuchi
- BioMedical Laboratory, Department of Research, Kitasato Institute Hospital, Tokyo, 108-8642, Japan
| | - Kazuhiro Watanabe
- Center for Education and Research on Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Masayoshi Hirohara
- Center for Education and Research on Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Yutaka Masuda
- Center for Education and Research on Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| |
Collapse
|
13
|
Boisteau E, Dahan L, Williet N, Le Malicot K, Desramé J, Bouché O, Petorin C, Malka D, Rebischung C, Aparicio T, Lecaille C, Rinaldi Y, Turpin A, Bignon AL, Bachet JB, Lepage C, Granger V, Legoux JL, Deplanque G, Baconnier M, Lecomte T, Bonnet I, Seitz JF, François E, Lièvre A. Clinico-biological factors predicting the benefit of the LV5FU2 maintenance strategy as a first-line therapy in patients with metastatic pancreatic cancer. Oncologist 2024; 29:e1149-e1158. [PMID: 39235326 PMCID: PMC11379635 DOI: 10.1093/oncolo/oyae079] [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: 12/02/2023] [Accepted: 03/13/2024] [Indexed: 09/06/2024] Open
Abstract
INTRODUCTION Predictive markers of LV5FU2 maintenance benefit after first-line induction with FOLFIRINOX in patients with metastatic pancreatic cancer are necessary to select patients who will not be harmed by this strategy. PATIENTS AND METHODS We focused on patients who received 12 cycles of FOLFIRINOX (arm A, N = 88) or 8 cycles of FOLFIRINOX followed by LV5FU2 maintenance in controlled patients (arm B, N = 91) from the PRODIGE-35 trial. Prognostic factors and predictors of efficiency were identified by using Cox regression. Median progression-free survival (PFS), overall survival (OS), and time to deterioration of quality of life (TTD-QoL) were evaluated. RESULTS Poor independent prognostic factors were primary tumor in place, age <65 years and the presence of liver metastases for PFS, a baseline neutrophil/lymphocyte ratio (NLR) ≥5 and CA19.9 ≥500 UI/L for OS, independent of the treatment arm. Patients with one metastatic site had a longer PFS in arm A, whereas patients with ≥2 metastatic sites had a longer PFS in arm B. We also identified predictors of OS and TTD-QoL in arm B but these differences were not statistically significant. CONCLUSION Except for patients with one metastatic site who benefited more from 12 cycles of FOLFIRINOX, a maintenance strategy with LV5FU2 should be widely offered to mPC patients whose survival and QoL are preserved after 4 months of FOLFIRINOX. (ClinicalTrials.gov: NCT02352337).
Collapse
Affiliation(s)
- Emeric Boisteau
- Department of Gastroenterology, University Hospital Pontchaillou, Rennes 1 University, INSERM U1242 "Chemistry Oncogenesis Stress Signalling," Rennes, France
| | - Laetitia Dahan
- Department of Digestive Oncology, La Timone, AMU, Marseille, France
| | - Nicolas Williet
- Hepatogastroenterology Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Karine Le Malicot
- Biostatistics Department, Francophone Federation of Digestive Cancerology, EPICAD INSERM LNC-UMR 1231 University of Burgundy and Franche Comté, Dijon, France
| | | | - Olivier Bouché
- Department of Digestive Oncology, CHU Reims, Reims, France
| | - Caroline Petorin
- Department of Digestive Oncology, University Hospital d'Estaing, Clermont-Ferrand, France
| | - David Malka
- Department of Hepato-Gastroenterology, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Thomas Aparicio
- Department of Gastroenterology, Saint Louis Hospital, APHP, Université de Paris, Paris, France
| | | | - Yves Rinaldi
- Department of Hepato-gastroenterology, European Hospital, Marseille, France
| | - Anthony Turpin
- Univ. Lille, CNRS UMR9020, Inserm UMR-S 1277, Canther, Cancer Heterogeneity, Plasticity and Resistance to Therapies, CHU Lille, Department of Medical Oncology, Lille, France
| | - Anne-Laure Bignon
- Department of Digestive Oncology, University Hospital Côte de Nacre, Caen, France
| | - Jean-Baptiste Bachet
- Department of Hepato-gastroenterology, Pitié-Salpêtrière Hospital, Paris, France
| | - Côme Lepage
- Department of Hepato-gastroenterology, University hospital of Dijon, EPICAD INSERM LNC-UMR 1231, University of Burgundy and Franche Comté, Dijon, France
| | - Victoire Granger
- Department of Hepato-gastroenterology and Digestive Oncology, University Hospital of Grenoble Alpes, Grenoble, France
| | - Jean-Louis Legoux
- Department of Hepato-gastroenterology and Digestive Oncology, Orléans Regional Hospital, Orléans, France
| | - Gaël Deplanque
- Department of Hepato-Gastroenterology, Saint-Joseph Hospital, Paris, France
| | - Mathieu Baconnier
- Department of Hepato-Gastroenterology, Annecy-Genevois Hospital Center, Epagny Metz-Tessy, France
| | - Thierry Lecomte
- Department of Hepato-Gastroenterology and Digestive Oncology, University Hospital of Tours; UMR INSERM 1069 Nutrition, Croissance et Cancer, University of Tours, Tours, France
| | | | | | - Eric François
- Antoine Lacassagne Center, Oncology Department, Nice, France
| | - Astrid Lièvre
- Department of Gastroenterology, University Hospital Pontchaillou, Rennes 1 University, INSERM U1242 "Chemistry Oncogenesis Stress Signalling," Rennes, France
| |
Collapse
|
14
|
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
|
15
|
Cheng HH, Shevach JW, Castro E, Couch FJ, Domchek SM, Eeles RA, Giri VN, Hall MJ, King MC, Lin DW, Loeb S, Morgan TM, Offit K, Pritchard CC, Schaeffer EM, Szymaniak BM, Vassy JL, Katona BW, Maxwell KN. BRCA1, BRCA2, and Associated Cancer Risks and Management for Male Patients: A Review. JAMA Oncol 2024; 10:1272-1281. [PMID: 39052257 DOI: 10.1001/jamaoncol.2024.2185] [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] [Indexed: 07/27/2024]
Abstract
Importance Half of all carriers of inherited cancer-predisposing variants in BRCA1 and BRCA2 are male, but the implications for their health are underrecognized compared to female individuals. Germline variants in BRCA1 and BRCA2 (also known as pathogenic or likely pathogenic variants, referred to here as BRCA1/2 PVs) are well known to significantly increase the risk of breast and ovarian cancers in female carriers, and knowledge of BRCA1/2 PVs informs established cancer screening and options for risk reduction. While risks to male carriers of BRCA1/2 PVs are less characterized, there is convincing evidence of increased risk for prostate cancer, pancreatic cancer, and breast cancer in males. There has also been a rapid expansion of US Food and Drug Administration-approved targeted cancer therapies, including poly ADP ribose polymerase (PARP) inhibitors, for breast, pancreatic, and prostate cancers associated with BRCA1/2 PVs. Observations This narrative review summarized the data that inform cancer risks, targeted cancer therapy options, and guidelines for early cancer detection. It also highlighted areas of emerging research and clinical trial opportunities for male BRCA1/2 PV carriers. These developments, along with the continued relevance to family cancer risk and reproductive options, have informed changes to guideline recommendations for genetic testing and strengthened the case for increased genetic testing for males. Conclusions and Relevance Despite increasing clinical actionability for male carriers of BRCA1/2 PVs, far fewer males than female individuals undergo cancer genetic testing. Oncologists, internists, and primary care clinicians should be vigilant about offering appropriate genetic testing to males. Identifying more male carriers of BRCA1/2 PVs will maximize opportunities for cancer early detection, targeted risk management, and cancer treatment for males, along with facilitating opportunities for risk reduction and prevention in their family members, thereby decreasing the burden of hereditary cancer.
Collapse
Affiliation(s)
- Heather H Cheng
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Medicine (Hematology and Oncology), University of Washington, Seattle
| | - Jeffrey W Shevach
- Division of Medical Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Elena Castro
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fergus J Couch
- Division of Experimental Pathology and Laboratory Medicine, Mayo Clinic, Rochester, New York
| | - Susan M Domchek
- Department of Medicine, Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Rosalind A Eeles
- The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Veda N Giri
- Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut
| | - Michael J Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Mary-Claire King
- Department of Medicine (Medical Genetics) and Department of Genome Sciences, University of Washington, Seattle
| | - Daniel W Lin
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Urology, University of Washington, Seattle
| | - Stacy Loeb
- Department of Urology and Population Health, New York University School of Medicine, New York
- Department of Surgery/Urology, Manhattan Veterans Affairs, New York, New York
| | - Todd M Morgan
- Department of Urology, University of Michigan, Ann Arbor
| | - Kenneth Offit
- Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Colin C Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
- Brotman Baty Institute for Precision Medicine, Seattle, Washington
| | - Edward M Schaeffer
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Brittany M Szymaniak
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jason L Vassy
- Harvard Medical School at VA Boston Healthcare System, Boston, Massachusetts
| | - Bryson W Katona
- Department of Medicine, Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Kara N Maxwell
- Department of Medicine, Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Philadelphia
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania
| |
Collapse
|
16
|
Kawamoto Y, Yamai T, Ikezawa K, Seiki Y, Watsuji K, Hirao T, Urabe M, Kai Y, Takada R, Mukai K, Nakabori T, Uehara H, Inoue T, Fujisawa F, Ohkawa K. Clinical significance of germline breast cancer susceptibility gene (gBRCA) testing and olaparib as maintenance therapy for patients with pancreatic cancer. BMC Cancer 2024; 24:1000. [PMID: 39134950 PMCID: PMC11321060 DOI: 10.1186/s12885-024-12722-8] [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: 02/10/2024] [Accepted: 07/29/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Germline breast cancer susceptibility gene (gBRCA) mutation in patients with pancreatic cancer (PC) is not common in clinical practice. Therefore, factors that efficiently show gBRCA mutations and the real-world outcomes of olaparib maintenance therapy have not been fully established. In the present study, we clarified the indicators for the effective detection of gBRCA mutation and the efficacy and safety of olaparib as maintenance therapy. METHODS We retrospectively analyzed 84 patients with PC who underwent gBRCA testing (BRACAnalysis, Myriad Genetics, Salt Lake City, UT, USA) at our institute between January 2021 and March 2022. For each patient, clinical data were extracted from medical records. RESULTS The median patient age was 64 y (29-85 y), and 41 patients (48.8%) were male. The gBRCA mutations were identified in 10 (11.9%) patients; two patients had BRCA1 mutation and eight had BRCA2 mutation. All patients with gBRCA mutation had a family history of any cancer, and eight of them had a family history of Hereditary Breast and Ovarian Cancer syndrome (HBOC)-related cancer. The gBRCA mutation rate was higher for patients with PC with a family history of HBOC-related cancer compared to that in patients with PC having a family history of other cancers and no family history of cancer (22.9% vs. 4.1%; P = 0.014). In our study, eight out of 10 patients with gBRCA-positive PC received olaparib after platinum-based chemotherapy. The best responses to platinum-based chemotherapy included a complete response in one patient (12.5%) and a partial response in seven patients (87.5%). The median duration of treatment with platinum-based chemotherapy plus olaparib was 17.5 months (8-87 months), and the duration of treatment with olaparib maintenance therapy was 11 months (1-30 months). During olaparib maintenance therapy, three patients showed no disease progression. One of these three patients underwent conversion surgery after receiving olaparib for 12 months. CONCLUSIONS The gBRCA testing should be considered proactively, especially in patients with PC with a family history of HBOC-related cancer.
Collapse
Affiliation(s)
- Yasuharu Kawamoto
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Takuo Yamai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
- Department of Gastroenterology, Osaka General Medical Center, Osaka, Japan
| | - Kenji Ikezawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan.
| | - Yusuke Seiki
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Ko Watsuji
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Takeru Hirao
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Makiko Urabe
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Yugo Kai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Ryoji Takada
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Kaori Mukai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Tasuku Nakabori
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Hiroyuki Uehara
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Tazuko Inoue
- Department of Genetic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Fumie Fujisawa
- Department of Genetic Oncology, Osaka International Cancer Institute, Osaka, Japan
- Department of Medical Oncology, Shiga General Hospital, Shiga, Japan
| | - Kazuyoshi Ohkawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| |
Collapse
|
17
|
Bazan Russo TD, Mujacic C, Di Giovanni E, Vitale MC, Ferrante Bannera C, Randazzo U, Contino S, Bono M, Gristina V, Galvano A, Perez A, Badalamenti G, Russo A, Bazan V, Incorvaia L. Polθ: emerging synthetic lethal partner in homologous recombination-deficient tumors. Cancer Gene Ther 2024:10.1038/s41417-024-00815-2. [PMID: 39122831 DOI: 10.1038/s41417-024-00815-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024]
Abstract
The most remarkable finding in synthetic lethality (SL) is the hypersensitivity to PARP inhibitors (PARPis) of the tumors harboring defects in genes involved in homologous repair (HR) such as BRCA1/2. Despite initial responsiveness to PARPi, the penetrance of the synthetic lethal interactions between BRCA1/2 genes and PARPi is incomplete. Thus, a significant proportion of HR-defective tumors experience intrinsic or acquired resistance, representing a key challenge of clinical research. An expanded concept of SL is opening new ways and includes novel forms of genetic interactions, investigating not only traditional SL of pairs genes but also SL between biological pathways that regulate the same essential survival cell function. In this context, recent research showed that HR and theta-mediated end-joining (TMEJ) pathways exhibit SL. DNA polymerase theta (Polθ) is encoded by the POLQ gene and is a key component of the TMEJ, an essential backup pathway, intrinsically mutagenic, to repair resected double-strand breaks (DSBs) when the non-homologous end joining (NHEJ) and HR are impaired. Polθ is broadly expressed in normal tissues, overexpressed in several cancers, and typically associated with poor outcomes and shorter relapse-free survival. Notably, HR-deficient tumor cells present the characteristic mutational signatures of the error-prone TMEJ pathway. According to this observation, the loss of HR proteins, such as BRCA1 or BRCA2, contributes to increasing the TMEJ-specific genomic profile, suggesting synthetic lethal interactions between loss of the POLQ and HR genes, and resulting in the emerging interest for Polθ as a potential therapeutic target in BRCA1/2-associated tumors.This review summarizes the converging roles of the POLQ and HR genes in DNA DSB repair, the early-stage clinical trials using Polθ inhibitor to treat HR-defective tumors and to overcome BRCA-reversion mutations responsible for therapeutic resistance, and the novel pleiotropic effects of Polθ, paving the way for the development of unexplored synthetic lethality strategies.
Collapse
Affiliation(s)
- Tancredi Didier Bazan Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Clarissa Mujacic
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Emilia Di Giovanni
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Maria Concetta Vitale
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Carla Ferrante Bannera
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Ugo Randazzo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Silvia Contino
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Marco Bono
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Valerio Gristina
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Antonio Galvano
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Alessandro Perez
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Giuseppe Badalamenti
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy.
| | - Antonio Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy.
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Lorena Incorvaia
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| |
Collapse
|
18
|
Mohan A, Quingalahua E, Gunchick V, Paul S, Kumar-Sinha C, Crysler O, Zalupski MM, Sahai V. PARP inhibitor therapy in patients with IDH1 mutated cholangiocarcinoma. Oncologist 2024; 29:725-730. [PMID: 39036962 PMCID: PMC11299928 DOI: 10.1093/oncolo/oyae163] [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: 06/06/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Isocitrate dehydrogenase 1 (IDH1) missense mutations occur at a frequency of 10%-15% in intrahepatic cholangiocarcinoma (iCCA). IDH1 mutations result in accumulation of (R)-2-hydroxyglutarate, an oncometabolite that leads to DNA hypermethylation and impairment of homologous recombination (HR). Impairment of HR results in a "BRCAness" phenotype which may confer sensitivity to poly(ADP ribose) polymerase (PARP) inhibition. METHODS We conducted a retrospective cohort review to identify patients with advanced, IDH1 mutated iCCA treated with a PARP inhibitor (PARPi) at the University of Michigan between 2018 and 2023. Patients are described with respect to prior lines of therapy, response to platinum-based chemotherapy, and progression-free survival (PFS) and overall survival (OS) from the time of PARPi initiation. RESULTS Between 2018 and 2023 we identified 40 patients with IDH1 mutated iCCA of which 6 patients were treated with a PARPi as monotherapy or in combination with an ATR inhibitor or anti-PD-1 immune checkpoint inhibitor. Majority of patients (n = 5) carried an IDH1 R132C mutation per tissue-based next generation sequencing. All patients had previously received at least one line of cisplatin-based systemic therapy for advanced disease prior to treatment with PARPi. PFS and OS from time of PARPi initiation ranged from 1.4 to 18.5 months and 2.8 to 42.4 months, respectively. Best response on PARPi therapy included 2 partial responses. CONCLUSION This is the first case series to describe PARPi treatment in IDH1 mutated iCCA. Results underscore the limitation of PARPi monotherapy, potentially support combined PARPi therapies, and highlight a need for effective treatment options for patients with IDH1 mutated iCCA.
Collapse
Affiliation(s)
- Arathi Mohan
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Elit Quingalahua
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Valerie Gunchick
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Simi Paul
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Chandan Kumar-Sinha
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, United States
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Oxana Crysler
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Mark M Zalupski
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Vaibhav Sahai
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
19
|
Urabe M, Ikezawa K, Kozumi K, Kai Y, Takada R, Mukai K, Nakabori T, Uehara H, Akita H, Ohkawa K. Long-term survival after systemic chemotherapy, chemoradiotherapy, and maintenance therapy for an older adult patient with recurrent pancreatic acinar cell carcinoma. Clin J Gastroenterol 2024; 17:771-775. [PMID: 38743170 DOI: 10.1007/s12328-024-01981-4] [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: 02/06/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024]
Abstract
Pancreatic acinar cell carcinoma (PACC) is a rare cancer with no specific treatment. The treatment and chemotherapy for PACC are selected according to pancreatic ductal adenocarcinoma (PDAC). Herein, we describe a recurrent PACC case of an older adult patient. The patient was treated with systemic chemotherapy, chemoradiotherapy, and maintenance therapy based on the pathologic germline BRCA2 variant, resulting in long-term survival. The pathogenic BRCA variant is detected more frequently in patients with PACC than in those with PDAC. The BRCA variant significantly impacts treatment selection and prognosis; therefore, early genomic analysis is recommended when treating PACC.
Collapse
Affiliation(s)
- Makiko Urabe
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Kenji Ikezawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan.
| | - Kazuhiro Kozumi
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Yugo Kai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Ryoji Takada
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Kaori Mukai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Tasuku Nakabori
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Hiroyuki Uehara
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Hirofumi Akita
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuyoshi Ohkawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| |
Collapse
|
20
|
Lebedeva A, Veselovsky E, Kavun A, Belova E, Grigoreva T, Orlov P, Subbotovskaya A, Shipunov M, Mashkov O, Bilalov F, Shatalov P, Kaprin A, Shegai P, Diuzhev Z, Migiaev O, Vytnova N, Mileyko V, Ivanov M. Untapped Potential of Poly(ADP-Ribose) Polymerase Inhibitors: Lessons Learned From the Real-World Clinical Homologous Recombination Repair Mutation Testing. World J Oncol 2024; 15:562-578. [PMID: 38993246 PMCID: PMC11236374 DOI: 10.14740/wjon1820] [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: 01/17/2024] [Accepted: 04/29/2024] [Indexed: 07/13/2024] Open
Abstract
Background Testing for homologous recombination deficiency (HRD) mutations is pivotal to assess individual risk, to proact preventive measures in healthy carriers and to tailor treatments for cancer patients. Increasing prominence of poly(ADP-ribose) polymerase (PARP) inhibitors with remarkable impact on molecular-selected patient survival across diverse nosologies, ingrains testing for BRCA genes and beyond in clinical practice. Nevertheless, testing strategies remain a question of debate. While several pathogenic BRCA1/2 gene variants have been described as founder pathogenic mutations frequently found in patients from Russia, other homologous recombination repair (HRR) genes have not been sufficiently explored. In this study, we present real-world data of routine HRR gene testing in Russia. Methods We evaluated clinical and sequencing data from cancer patients who had germline/somatic next-generation sequencing (NGS) HRR gene testing in Russia (BRCA1/2/ATM/CHEK2, or 15 HRR genes). The primary objectives of this study were to evaluate the frequency of BRCA1/2 and non-BRCA gene mutations in real-world unselected patients from Russia, and to determine whether testing beyond BRCA1/2 is feasible. Results Data of 2,032 patients were collected from February 2021 to February 2023. Most had breast (n = 715, 35.2%), ovarian (n = 259, 12.7%), pancreatic (n = 85, 4.2%), or prostate cancer (n = 58, 2.9%). We observed 586 variants of uncertain significance (VUS) and 372 deleterious variants (DVs) across 487 patients, with 17.6% HRR-mutation positivity. HRR testing identified 120 (11.8%) BRCA1/2-positive, and 172 (16.9%) HRR-positive patients. With 51 DVs identified in 242 formalin-fixed paraffin-embedded (FFPE), testing for variant origin clarification was required in one case (0.4%). Most BRCA1/2 germline variants were DV (121 DVs, 26 VUS); in non-BRCA1/2 genes, VUS were ubiquitous (53 DVs, 132 VUS). In silico prediction identified additional 4.9% HRR and 1.2% BRCA1/2/ATM/CHEK2 mutation patients. Conclusions Our study represents one of the first reports about the incidence of DV and VUS in HRR genes, including genes beyond BRCA1/2, identified in cancer patients from Russia, assessed by NGS. In silico predictions of the observed HRR gene variants suggest that non-BRCA gene testing is likely to result in higher frequency of patients who are candidates for PARP inhibitor therapy. Continuing sequencing efforts should clarify interpretation of frequently observed non-BRCA VUS.
Collapse
Affiliation(s)
- Alexandra Lebedeva
- OncoAtlas LLC, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Egor Veselovsky
- OncoAtlas LLC, Moscow, Russia
- Department of Evolutionary Genetics of Development, Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | | | - Ekaterina Belova
- OncoAtlas LLC, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Tatiana Grigoreva
- OncoAtlas LLC, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Pavel Orlov
- The Federal Research Center for Fundamental and Translational Medicine (NIIECM FRC FTM), Novosibirsk, Russia
| | - Anna Subbotovskaya
- The Federal Research Center for Fundamental and Translational Medicine (NIIECM FRC FTM), Novosibirsk, Russia
| | - Maksim Shipunov
- The Federal Research Center for Fundamental and Translational Medicine (NIIECM FRC FTM), Novosibirsk, Russia
| | - Oleg Mashkov
- State Budgetary Institution of Healthcare Republican Medical Genetic Center, Ufa, Russia
| | - Fanil Bilalov
- State Budgetary Institution of Healthcare Republican Medical Genetic Center, Ufa, Russia
| | - Peter Shatalov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Andrey Kaprin
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Peter Shegai
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | | | | | | | - Vladislav Mileyko
- OncoAtlas LLC, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maxim Ivanov
- OncoAtlas LLC, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia
| |
Collapse
|
21
|
Yavas A, Ozcan K, Adsay NV, Balci S, Tarcan ZC, Hechtman JF, Luchini C, Scarpa A, Lawlor RT, Mafficini A, Reid MD, Xue Y, Yang Z, Haye K, Bellizzi AM, Vanoli A, Benhamida J, Balachandran V, Jarnagin W, Park W, O'Reilly EM, Klimstra DS, Basturk O. SWI/SNF Complex-Deficient Undifferentiated Carcinoma of the Pancreas: Clinicopathologic and Genomic Analysis. Mod Pathol 2024; 37:100585. [PMID: 39094734 DOI: 10.1016/j.modpat.2024.100585] [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: 04/08/2024] [Revised: 07/12/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
Inactivating alterations in the SWItch/Sucrose NonFermentable (SWI/SNF) Chromatin Remodeling Complex subunits have been described in multiple tumor types. Recent studies focused on SMARC subunits of this complex to understand their relationship with tumor characteristics and therapeutic opportunities. To date, pancreatic cancer with these alterations has not been well studied, although isolated cases of undifferentiated carcinomas have been reported. Herein, we screened 59 pancreatic undifferentiated carcinomas for alterations in SWI/SNF complex-related (SMARCB1 [BAF47/INI1], SMARCA4 [BRG1], SMARCA2 [BRM]) proteins and/or genes using immunohistochemistry and/or next-generation sequencing. Cases with alterations in SWI/SNF complex-related proteins/genes were compared with cases without alterations, as well as with 96 conventional pancreatic ductal adenocarcinomas (PDAC). In all tumor groups, mismatch repair and PD-L1 protein expression were also evaluated. Thirty of 59 (51%) undifferentiated carcinomas had a loss of SWI/SNF complex-related protein expression or gene alteration. Twenty-seven of 30 (90%) SWI-/SNF-deficient undifferentiated carcinomas had rhabdoid morphology (vs 9/29 [31%] SWI-/SNF-retained undifferentiated carcinomas; P < .001) and all expressed cytokeratin, at least focally. Immunohistochemically, SMARCB1 protein expression was absent in 16/30 (53%) cases, SMARCA2 in 4/30 (13%), and SMARCA4 in 4/30 (13%); both SMARCB1 and SMARCA2 protein expressions were absent in 1/30 (3%). Five of 8 (62.5%) SWI-/SNF-deficient undifferentiated carcinomas that displayed loss of SMARCB1 protein expression by immunohistochemistry were found to have corresponding SMARCB1 deletions by next-generation sequencing. Analysis of canonical driver mutations for PDAC in these cases showed KRAS (2/5) and TP53 (2/5) abnormalities. Median combined positive score for PD-L1 (E1L3N) was significantly higher in the undifferentiated carcinomas with/without SWI/SNF deficiency compared with the conventional PDACs (P < .001). SWI-/SNF-deficient undifferentiated carcinomas were larger (P < .001) and occurred in younger patients (P < .001). Patients with SWI-/SNF-deficient undifferentiated carcinoma had worse overall survival compared with patients with SWI-/SNF-retained undifferentiated carcinoma (P = .004) and PDAC (P < .001). Our findings demonstrate that SWI-/SNF-deficient pancreatic undifferentiated carcinomas are frequently characterized by rhabdoid morphology, exhibit highly aggressive behavior, and have a negative prognostic impact. The ones with SMARCB1 deletions appear to be frequently KRAS wild type. Innovative developmental therapeutic strategies targeting this genomic basis of the SWI/SNF complex and the therapeutic implications of EZH2 inhibition (NCT03213665), SMARCA2 degrader (NCT05639751), or immunotherapy are currently under investigation.
Collapse
Affiliation(s)
- Aslihan Yavas
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pathology, Now with Institute of Pathology, Heinrich Heine University and University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Kerem Ozcan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Now with Department of Pathology and Laboratory Medicine, Henry Ford Hospital, Detroit, Michigan
| | - N Volkan Adsay
- The Department of Pathology, Koç University Hospital and Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Serdar Balci
- Department of Pathology, Memorial Healthcare Group, Istanbul, Turkey
| | - Zeynep C Tarcan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; David M. Rubenstein Center for Pancreatic Cancer Research, New York, New York
| | - Jaclyn F Hechtman
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Now with Caris Life Sciences, Miami, Florida
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy; ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Rita T Lawlor
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, Verona, Italy
| | - Andrea Mafficini
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, Verona, Italy
| | - Michelle D Reid
- Department of Pathology, School of Medicine, Emory University, Atlanta, Georgia
| | - Yue Xue
- Department of Pathology, University Hospitals, Cleveland, Ohio
| | - Zhaohai Yang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kester Haye
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Andrew M Bellizzi
- Department of Pathology, University of Iowa, Iowa City, Iowa; Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, Pavia, Italy
| | - Alessandro Vanoli
- Unit of Anatomic Pathology, Fondazione IRCCS San Matteo Hospital, Pavia, Italy
| | - Jamal Benhamida
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vinod Balachandran
- David M. Rubenstein Center for Pancreatic Cancer Research, New York, New York; Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William Jarnagin
- David M. Rubenstein Center for Pancreatic Cancer Research, New York, New York; Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Wungki Park
- David M. Rubenstein Center for Pancreatic Cancer Research, New York, New York; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eileen M O'Reilly
- David M. Rubenstein Center for Pancreatic Cancer Research, New York, New York; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David S Klimstra
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Now with Paige.AI, New York, New York
| | - Olca Basturk
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; David M. Rubenstein Center for Pancreatic Cancer Research, New York, New York.
| |
Collapse
|
22
|
Dai MF, Wang X, Xin WX, Kong SS, Xu WB, Ding HY, Fang L. Safety and hematological toxicities of PARP inhibitors in patients with cancer: a systematic review of randomized controlled trials and a pharmacovigilance analysis. Expert Rev Anticancer Ther 2024; 24:613-622. [PMID: 38761169 DOI: 10.1080/14737140.2024.2357822] [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: 11/03/2023] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
INTRODUCTION This study aimed to estimate the toxicities of PARP inhibitors (PARPis), based on randomized controlled trials (RCTs) and the FDA Adverse Event Reporting System (FAERS) database. METHODS Four electronic databases were searched from inception to 16 April 2024, for RCTs of approved PARPis. The primary and secondary outcomes were grade 3-5 adverse events (AEs) and grade 3-5 hematological AE, respectively. We conducted network meta-analyses to calculate the relative risks (RRs) and 95% confidence intervals (CIs) of outcomes. A disproportionality analysis was conducted to estimate the signals of hematological AEs associated with PARPis from the FAERS database. RESULTS Overall, 27 RCTs involving 11,067 patients with cancer were included. Olaparib had the best safety profile for any grade 3-5 AEs and hematological AEs among four approved PARPis. Olaparib did not increase the risk of thrombocytopenia (RR: 1.48; 95%CI: 0.64-3.39), but other PARPis did. Furthermore 14,780 hematological AE reports associated with PARPis were identified in the FAERS database, and all PARPis were associated with strong hematological AE signals. Hematological AEs mainly occurred within the first 3 months (80.84%) after PARPi initiation. CONCLUSION Olaparib had the best safety profile among five PARPis. PARPi-associated hematological AEs mainly occurred within the first 3 months. REGISTRATION PROSPERO (CRD42022385274).
Collapse
Affiliation(s)
- Meng-Fei Dai
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xin Wang
- Department of Pharmacy, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Xiu Xin
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Si-Si Kong
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Wei-Ben Xu
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Hai-Ying Ding
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Luo Fang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
23
|
Deiana C, Agostini M, Brandi G, Giovannetti E. The trend toward more target therapy in pancreatic ductal adenocarcinoma. Expert Rev Anticancer Ther 2024; 24:525-565. [PMID: 38768098 DOI: 10.1080/14737140.2024.2357802] [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: 12/19/2023] [Accepted: 05/16/2024] [Indexed: 05/22/2024]
Abstract
INTRODUCTION Despite the considerable progress made in cancer treatment through the development of target therapies, pancreatic ductal adenocarcinoma (PDAC) continues to exhibit resistance to this category of drugs. As a result, chemotherapy combination regimens remain the primary treatment approach for this aggressive cancer. AREAS COVERED In this review, we provide an in-depth analysis of past and ongoing trials on both well-known and novel targets that are being explored in PDAC, including PARP, EGFR, HER2, KRAS, and its downstream and upstream pathways (such as RAF/MEK/ERK and PI3K/AKT/mTOR), JAK/STAT pathway, angiogenesis, metabolisms, epigenetic targets, claudin, and novel targets (such as P53 and plectin). We also provide a comprehensive overview of the significant trials for each target, allowing a thorough glimpse into the past and future of target therapy. EXPERT OPINION The path toward implementing a target therapy capable of improving the overall survival of PDAC is still long, and it is unlikely that a monotherapy target drug will fulfill a meaningful role in addressing the complexity of this cancer. Thus, we discuss the future direction of target therapies in PDAC, trying to identify the more promising target and combination treatments, with a special focus on the more eagerly awaited ongoing trials.
Collapse
Affiliation(s)
- Chiara Deiana
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Margherita Agostini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giovanni Brandi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Associazione Italiana per la Ricerca sul Cancro (AIRC) Start-Up Unit, Fondazione Pisana per la Scienza, Pisa, San Giuliano, Italy
| |
Collapse
|
24
|
Huang S, Yang J, Xie T, Jiang Y, Hong Y, Liu X, He X, Buratto D, Zhang D, Zhou R, Liang T, Bai X. Inhibition of DEF-p65 Interactions as a Potential Avenue to Suppress Tumor Growth in Pancreatic Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401845. [PMID: 38757623 PMCID: PMC11267266 DOI: 10.1002/advs.202401845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/28/2024] [Indexed: 05/18/2024]
Abstract
The limited success of current targeted therapies for pancreatic cancer underscores an urgent demand for novel treatment modalities. The challenge in mitigating this malignancy can be attributed to the digestive organ expansion factor (DEF), a pivotal yet underexplored factor in pancreatic tumorigenesis. The study uses a blend of in vitro and in vivo approaches, complemented by the theoretical analyses, to propose DEF as a promising anti-tumor target. Analysis of clinical samples reveals that high expression of DEF is correlated with diminished survival in pancreatic cancer patients. Crucially, the depletion of DEF significantly impedes tumor growth. The study further discovers that DEF binds to p65, shielding it from degradation mediated by the ubiquitin-proteasome pathway in cancer cells. Based on these findings and computational approaches, the study formulates a DEF-mimicking peptide, peptide-031, designed to disrupt the DEF-p65 interaction. The effectiveness of peptide-031 in inhibiting tumor proliferation has been demonstrated both in vitro and in vivo. This study unveils the oncogenic role of DEF while highlighting its prognostic value and therapeutic potential in pancreatic cancer. In addition, peptide-031 is a promising therapeutic agent with potent anti-tumor effects.
Collapse
Affiliation(s)
- Sicong Huang
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Key Laboratory of Pancreatic Disease of Zhejiang ProvinceHangzhou310000China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang ProvinceHangzhou310000China
| | - Jiaqi Yang
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Key Laboratory of Pancreatic Disease of Zhejiang ProvinceHangzhou310000China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang ProvinceHangzhou310000China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic DiseasesHangzhou310000China
| | - Teng Xie
- Institute of Quantitative Biology, College of Life SciencesZhejiang UniversityHangzhou310000China
- Shanghai Institute for Advanced StudyZhejiang UniversityShanghai200000China
| | - Yangwei Jiang
- Institute of Quantitative Biology, College of Life SciencesZhejiang UniversityHangzhou310000China
| | - Yifan Hong
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Key Laboratory of Pancreatic Disease of Zhejiang ProvinceHangzhou310000China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang ProvinceHangzhou310000China
| | - Xinyuan Liu
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Key Laboratory of Pancreatic Disease of Zhejiang ProvinceHangzhou310000China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang ProvinceHangzhou310000China
| | - Xuyan He
- Life Sciences InstituteZhejiang UniversityHangzhou310000China
| | - Damiano Buratto
- Institute of Quantitative Biology, College of Life SciencesZhejiang UniversityHangzhou310000China
- Shanghai Institute for Advanced StudyZhejiang UniversityShanghai200000China
| | - Dong Zhang
- Institute of Quantitative Biology, College of Life SciencesZhejiang UniversityHangzhou310000China
- Shanghai Institute for Advanced StudyZhejiang UniversityShanghai200000China
| | - Ruhong Zhou
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Institute of Quantitative Biology, College of Life SciencesZhejiang UniversityHangzhou310000China
- Shanghai Institute for Advanced StudyZhejiang UniversityShanghai200000China
- Department of ChemistryColumbia UniversityNew York10027USA
- Cancer CenterZhejiang UniversityHangzhou310000China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Key Laboratory of Pancreatic Disease of Zhejiang ProvinceHangzhou310000China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang ProvinceHangzhou310000China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic DiseasesHangzhou310000China
- Cancer CenterZhejiang UniversityHangzhou310000China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Key Laboratory of Pancreatic Disease of Zhejiang ProvinceHangzhou310000China
- Innovation Center for the Study of Pancreatic Diseases of Zhejiang ProvinceHangzhou310000China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic DiseasesHangzhou310000China
- Cancer CenterZhejiang UniversityHangzhou310000China
| |
Collapse
|
25
|
Mosele MF, Westphalen CB, Stenzinger A, Barlesi F, Bayle A, Bièche I, Bonastre J, Castro E, Dienstmann R, Krämer A, Czarnecka AM, Meric-Bernstam F, Michiels S, Miller R, Normanno N, Reis-Filho J, Remon J, Robson M, Rouleau E, Scarpa A, Serrano C, Mateo J, André F. Recommendations for the use of next-generation sequencing (NGS) for patients with advanced cancer in 2024: a report from the ESMO Precision Medicine Working Group. Ann Oncol 2024; 35:588-606. [PMID: 38834388 DOI: 10.1016/j.annonc.2024.04.005] [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: 02/20/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Advancements in the field of precision medicine have prompted the European Society for Medical Oncology (ESMO) Precision Medicine Working Group to update the recommendations for the use of tumour next-generation sequencing (NGS) for patients with advanced cancers in routine practice. METHODS The group discussed the clinical impact of tumour NGS in guiding treatment decision using the ESMO Scale for Clinical Actionability of molecular Targets (ESCAT) considering cost-effectiveness and accessibility. RESULTS As for 2020 recommendations, ESMO recommends running tumour NGS in advanced non-squamous non-small-cell lung cancer, prostate cancer, colorectal cancer, cholangiocarcinoma, and ovarian cancer. Moreover, it is recommended to carry out tumour NGS in clinical research centres and under specific circumstances discussed with patients. In this updated report, the consensus within the group has led to an expansion of the recommendations to encompass patients with advanced breast cancer and rare tumours such as gastrointestinal stromal tumours, sarcoma, thyroid cancer, and cancer of unknown primary. Finally, ESMO recommends carrying out tumour NGS to detect tumour-agnostic alterations in patients with metastatic cancers where access to matched therapies is available. CONCLUSION Tumour NGS is increasingly expanding its scope and application within oncology with the aim of enhancing the efficacy of precision medicine for patients with cancer.
Collapse
Affiliation(s)
- M F Mosele
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - C B Westphalen
- Comprehensive Cancer Center Munich & Department of Medicine III, University Hospital, LMU Munich, Munich
| | - A Stenzinger
- Institute of Pathology, University Hospital Heidelberg and Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - F Barlesi
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre
| | - A Bayle
- Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre; Drug Development Department (DITEP), Gustave Roussy, Villejuif; Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif; Service de Biostatistique et Epidémiologie, Gustave Roussy, Villejuif
| | - I Bièche
- Department of Genetics, Institut Curie, INSERM U1016, Université Paris Cité, Paris, France
| | - J Bonastre
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif; Service de Biostatistique et Epidémiologie, Gustave Roussy, Villejuif
| | - E Castro
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid
| | - R Dienstmann
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona; University of Vic-Central University of Catalonia, Vic, Spain; Oncoclínicas, São Paulo, Brazil
| | - A Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - A M Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw; Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - F Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - S Michiels
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif; Service de Biostatistique et Epidémiologie, Gustave Roussy, Villejuif
| | - R Miller
- Department of Medical Oncology, University College London, London; Department of Medical Oncology, St Bartholomew's Hospital, London, UK
| | - N Normanno
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - J Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - J Remon
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - M Robson
- Breast Medicine and Clinical Genetics Services, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - E Rouleau
- Tumor Genetics Service, Medical Biology and Pathology Department, Gustave Roussy, Villejuif, France
| | - A Scarpa
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona-School of Medicine, Verona, Italy
| | - C Serrano
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona
| | - J Mateo
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona
| | - F André
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre.
| |
Collapse
|
26
|
Rosso C, Marciano ND, Nathan D, Chen WP, McLaren CE, Osann KE, Flodman PL, Cho MT, Lee FC, Dayyani F, Zell JA, Valerin JB. Hereditary Cancer Clinics Improve Adherence to NCCN Germline Testing Guidelines for Pancreatic Cancer. J Natl Compr Canc Netw 2024; 22:299-305. [PMID: 38889755 PMCID: PMC11462954 DOI: 10.6004/jnccn.2023.7333] [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: 06/07/2023] [Accepted: 12/21/2023] [Indexed: 06/20/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis, with a 5-year overall survival rate of 10%. In November 2018, NCCN recommended that all patients with PDAC receive genetic counseling (GC) and germline testing regardless of family history. We hypothesized that patients with PDAC were more likely to be referred for testing after this change to the guidelines, regardless of presumed predictive factors, and that compliance would be further improved following the implementation of a hereditary cancer clinic (HCC). METHODS We conducted a single-institution retrospective analysis of patients diagnosed with PDAC from June 2017 through December 2021 at University of California, Irvine. We compared rates of genetics referral among patients in different diagnostic eras: the 18-month period before the NCCN Guideline change (pre-NCCN era: June 2017 through November 2018), 14 months following the change (post-NCCN era: December 2018 through January 2020), and 18 months after the creation of an HCC (HCC era: June 2020 through December 2021). Family and personal cancer history, genetics referral patterns, and results of GC were recorded. Data were compared using chi-square, Fisher exact, and multivariate analyses. RESULTS A total of 335 patients were treated for PDAC (123 pre-NCCN, 109 post-NCCN, and 103 HCC) at University of California, Irvine. Demographics across groups were comparable. Prior to the guideline changes, 30% were referred to GC compared with 54.7% in the post-NCCN era. After the implementation of the HCC, 77.4% were referred to GC (P<.0001). The odds ratio (OR) for referral to GC among patients with a positive family history of cancer progressively decreased following the change (pre-NCCN era: OR, 11.90 [95% CI, 3.00-80.14]; post-NCCN era: OR, 3.39 [95% CI, 1.13-10.76]; HCC era: OR, 3.11 [95% CI, 0.95-10.16]). CONCLUSIONS The 2018 updates to the NCCN Guidelines for PDAC recommending germline testing for all patients with PDAC significantly increased GC referral rates at our academic medical center. Implementation of an HCC further boosted compliance with guidelines.
Collapse
Affiliation(s)
- Claudia Rosso
- School of Medicine, University of California, Irvine, Irvine, CA
| | | | - Deepika Nathan
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California, Irvine, Irvine, CA
| | - Wen-Pin Chen
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
| | - Christine E. McLaren
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
| | - Kathryn E. Osann
- School of Medicine, University of California, Irvine, Irvine, CA
| | - Pamela L. Flodman
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California, Irvine, Irvine, CA
| | - May T. Cho
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
- Division of Hematology/Oncology, Department of Medicine, UCI School of Medicine, Irvine, CA
| | - Fa-Chyi Lee
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
- Division of Hematology/Oncology, Department of Medicine, UCI School of Medicine, Irvine, CA
| | - Farshid Dayyani
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
- Division of Hematology/Oncology, Department of Medicine, UCI School of Medicine, Irvine, CA
| | - Jason A. Zell
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
- Division of Hematology/Oncology, Department of Medicine, UCI School of Medicine, Irvine, CA
| | - Jennifer B. Valerin
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
- Division of Hematology/Oncology, Department of Medicine, UCI School of Medicine, Irvine, CA
| |
Collapse
|
27
|
Valdez BC, Tsimberidou AM, Yuan B, Nieto Y, Baysal MA, Chakraborty A, Andersen CR, Andersson BS. Synergistic cytotoxicity of histone deacetylase and poly-ADP ribose polymerase inhibitors and decitabine in pancreatic cancer cells: Implications for novel therapy. Oncotarget 2024; 15:361-373. [PMID: 38829622 PMCID: PMC11146633 DOI: 10.18632/oncotarget.28588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/17/2024] [Indexed: 06/05/2024] Open
Abstract
Histone deacetylase inhibitors (HDACi) can modulate the acetylation status of proteins, influencing the genomic instability exhibited by cancer cells. Poly (ADP ribose) polymerase (PARP) inhibitors (PARPi) have a direct effect on protein poly (ADP-ribosyl)ation, which is important for DNA repair. Decitabine is a nucleoside cytidine analogue, which when phosphorylated gets incorporated into the growing DNA strand, inhibiting methylation and inducing DNA damage by inactivating and trapping DNA methyltransferase on the DNA, thereby activating transcriptionally silenced DNA loci. We explored various combinations of HDACi and PARPi +/- decitabine (hypomethylating agent) in pancreatic cancer cell lines BxPC-3 and PL45 (wild-type BRCA1 and BRCA2) and Capan-1 (mutated BRCA2). The combination of HDACi (panobinostat or vorinostat) with PARPi (talazoparib or olaparib) resulted in synergistic cytotoxicity in all cell lines tested. The addition of decitabine further increased the synergistic cytotoxicity noted with HDACi and PARPi, triggering apoptosis (evidenced by increased cleavage of caspase 3 and PARP1). The 3-drug combination treatments (vorinostat, talazoparib, and decitabine; vorinostat, olaparib, and decitabine; panobinostat, talazoparib, and decitabine; panobinostat, olaparib, and decitabine) induced more DNA damage (increased phosphorylation of histone 2AX) than the individual drugs and impaired the DNA repair pathways (decreased levels of ATM, BRCA1, and ATRX proteins). The 3-drug combinations also altered the epigenetic regulation of gene expression (NuRD complex subunits, reduced levels). This is the first study to demonstrate synergistic interactions between the aforementioned agents in pancreatic cancer cell lines and provides preclinical data to design individualized therapeutic approaches with the potential to improve pancreatic cancer treatment outcomes.
Collapse
Affiliation(s)
- Benigno C. Valdez
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Apostolia M. Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bin Yuan
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mehmet A. Baysal
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Abhijit Chakraborty
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Clark R. Andersen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Borje S. Andersson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
28
|
Yu KH. Advances in Systemic Therapy in Pancreatic Cancer. Hematol Oncol Clin North Am 2024; 38:617-627. [PMID: 38575456 DOI: 10.1016/j.hoc.2024.03.002] [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: 04/06/2024]
Abstract
Substantial progress has been made toward understanding biology and developing new therapies for pancreatic ductal adenocarcinoma (PDAC). In this review, new insights from genomic profiling, as well as implications for treatment and prognosis, are discussed. New standards of care approaches with a focus on drug therapies are discussed for the treatment of resectable and advanced PDAC. The role of targeted and immune therapies remains limited; cohorts likely to benefit from these approaches are discussed. Promising, preliminary results regarding experimental therapies are reviewed.
Collapse
Affiliation(s)
- Kenneth H Yu
- Gastrointestinal Oncology Service, Cell Therapy Service, Memorial Sloan Kettering Cancer Center, 300 E 66th Street, New York, NY 10065, USA.
| |
Collapse
|
29
|
Wang L, Wulf GM. PAR caps the poles: a specific role for PARP2-inhibition to target the 'clustering' of extra spindle poles in cancer cells. EBioMedicine 2024; 104:105158. [PMID: 38749301 PMCID: PMC11108834 DOI: 10.1016/j.ebiom.2024.105158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024] Open
Affiliation(s)
- Lin Wang
- Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02215, USA
| | - Gerburg M Wulf
- Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02215, USA.
| |
Collapse
|
30
|
Srkalovic G, Rothe M, Mangat PK, Garrett-Mayer E, Ahn ER, Brouse G, Chan J, Mehmi I, Khalil M, Duvivier HL, Gaba A, Leuva H, Thota R, Yost KJ, Grantham GN, Gregory A, Hinshaw DC, Halabi S, Schilsky RL. Talazoparib in Patients With Solid Tumors With BRCA1/ 2 Mutation: Results From the Targeted Agent and Profiling Utilization Registry Study. JCO Precis Oncol 2024; 8:e2400026. [PMID: 38865672 DOI: 10.1200/po.24.00026] [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: 01/12/2024] [Revised: 02/22/2024] [Accepted: 04/04/2024] [Indexed: 06/14/2024] Open
Abstract
PURPOSE The Targeted Agent and Profiling Utilization Registry Study is a phase II basket trial evaluating the antitumor activity of commercially available targeted agents in patients with advanced cancer and genomic alterations known to be drug targets. Results of a cohort of patients with various solid tumors with germline or somatic BRCA1/2 mutations treated with talazoparib are reported. METHODS Eligible patients had advanced solid tumors, measurable disease (RECIST), Eastern Cooperative Oncology Group performance status 0-2, adequate organ function, and no standard treatment options. Patients with germline BRCA-mutated human epidermal growth factor receptor 2-negative locally advanced or metastatic breast cancer were not eligible for this study. Primary end point was disease control (DC) determined by investigator assessment of objective response (OR) or stable disease (SD) of at least 16 weeks duration (SD16+). The results were evaluated on the basis of a one-sided exact binomial test with a null DC rate of 15% versus 35% (power = 0.82; α = .10). Secondary end points were OR, progression-free survival, overall survival, duration of response, duration of SD, and safety. RESULTS Twenty-eight patients (20 cancer types) with BRCA1/2 mutations were enrolled from December 2019 to September 2021 and collapsed into a single histology pooled cohort for analysis. All patients were evaluable for efficacy. One complete response, nine partial response, and six SD16+ were observed for DC and OR rates of 57% (one-sided 90% CI, 43 to 100) and 36% (95% CI, 19 to 56), respectively. The null hypothesis of a 15% DC rate was rejected (P < .001). Patients with OR had the following tumor types: breast (2), nonmelanoma skin, mesothelioma, stomach, uterus, non-small cell lung cancer, ovary, hepatocellular carcinoma, and pancreas. Thirteen patients had at least one grade 3-5 adverse event (AE) or serious AE at least possibly related to talazoparib. All were consistent with the drug label except bilirubin increase and hyponatremia (both grade 3 AEs). CONCLUSION Talazoparib demonstrated antitumor activity in patients with advanced solid tumors and BRCA1/2 mutations, including cancer types for which poly ADP-ribose polymerase inhibitors are not yet US Food and Drug Administration-approved.
Collapse
Affiliation(s)
- Gordan Srkalovic
- Herbert-Herman Cancer Center, Lansing, MI
- Michigan Cancer Research Consortium, Ypsilanti, MI
| | - Michael Rothe
- American Society of Clinical Oncology, Alexandria, VA
| | - Pam K Mangat
- American Society of Clinical Oncology, Alexandria, VA
| | | | | | | | - John Chan
- Sutter Cancer Research Consortium, San Francisco, CA
| | - Inderjit Mehmi
- The Angeles Clinic and Research Institute, A Cedars-Sinai Affiliate, Los Angeles, CA
| | - Maya Khalil
- O'Neal Comprehensive Cancer Center at the University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | | | | | | | | | - Kathleen J Yost
- Cancer Research Consortium of West Michigan, Grand Rapids, MI
| | | | | | | | | | | |
Collapse
|
31
|
Ando Y, Dbouk M, Yoshida T, Abou Diwan E, Saba H, Dbouk A, Yoshida K, Roberts NJ, Klein AP, Burkhart R, He J, Hruban RH, Goggins M. Germline Pathogenic Variants in Patients With Pancreatic and Periampullary Cancers. JCO Precis Oncol 2024; 8:e2400101. [PMID: 38781545 DOI: 10.1200/po.24.00101] [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: 02/15/2024] [Revised: 03/11/2024] [Accepted: 04/02/2024] [Indexed: 05/25/2024] Open
Abstract
PURPOSE Inherited cancer susceptibility is often not suspected in the absence of a significant cancer family history. Pathogenic germline variants in pancreatic cancer are well-studied, and routine genetic testing is recommended in the guidelines. However, data on rare periampullary cancers other than pancreatic cancer are insufficient. We compared the prevalence of germline susceptibility variants in patients with pancreatic cancer and nonpancreatic periampullary cancers. MATERIALS AND METHODS Six hundred and eight patients who had undergone pancreaticoduodenal resection at a tertiary referral hospital were studied, including 213 with pancreatic ductal adenocarcinoma, 172 with ampullary cancer, 154 with distal common bile duct cancer, and 69 with duodenal adenocarcinoma. Twenty cancer susceptibility and candidate susceptibility genes were sequenced, and variant interpretation was assessed by interrogating ClinVar and PubMed. RESULTS Pathogenic or likely pathogenic, moderate- to high-penetrant germline variants were identified in 46 patients (7.7%), including a similar percentage of patients with pancreatic (8.5%) and nonpancreatic periampullary cancer (7.1%). Low-penetrant variants were identified in an additional 11 patients (1.8%). Eighty-nine percent of the moderate- to high-penetrant variants involved the major cancer susceptibility genes BRCA2, ATM, BRCA1, CDKN2A, MSH2/MLH1, and PALB2; the remaining 11% involved other cancer susceptibility genes such as BRIP1, BAP1, and MSH6. Almost all pathogenic variant carriers had a family history of cancer. CONCLUSION Patients with pancreatic and nonpancreatic periampullary cancer have a similar prevalence of pathogenic cancer susceptibility variants. Germline susceptibility testing should be considered for patients with any periampullary cancer.
Collapse
Affiliation(s)
- Yohei Ando
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Mohamad Dbouk
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Takeichi Yoshida
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Elizabeth Abou Diwan
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Helena Saba
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Ali Dbouk
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Kanako Yoshida
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Nicholas J Roberts
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Departments of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Alison P Klein
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Departments of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- The Bloomberg School of Public Health, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Richard Burkhart
- Departments of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Jin He
- Departments of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Ralph H Hruban
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Departments of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- The Bloomberg School of Public Health, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Michael Goggins
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Departments of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Departments of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|
32
|
Tentes AA, Kyziridis D, Kalakonas A, Iliadis A, Fotiadou A. Pancreatic cancer with synchronous peritoneal and hepatic metastases: A case report. Int J Surg Case Rep 2024; 118:109588. [PMID: 38581939 PMCID: PMC11015438 DOI: 10.1016/j.ijscr.2024.109588] [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: 02/14/2024] [Revised: 03/22/2024] [Accepted: 03/23/2024] [Indexed: 04/08/2024] Open
Abstract
Introduction and importance: There is evidence that patients with limited peritoneal carcinomatosis of pancreatic cancer or those with low burden of hepatic metastases are amenable to surgical resection. A case report of a patient with cancer of the pancreatic tail and synchronous peritoneal and hepatic metastases is presented. CASE PRESENTATION A male patient, 66 years old, underwent cytoreductive surgery (CRS) plus hyperthermic intraperitoneal chemotherapy (HIPEC) and radio-frequency ablation (RFA) for synchronous hepatic metastases simultaneously to distal pancreatectomy for adenocarcinoma of the pancreas. Adjuvant chemotherapy followed the R0 surgery. The patient remained disease free for 18 months, developed liver recurrence and died 28 months after the initial operation. DISCUSSION CRS plus HIPEC with synchronous ablation or resection of hepatic metastases may be used for the treatment of pancreatic cancer with synchronous peritoneal and hepatic metastases in highly selected patients. CONCLUSION Further studies are needed to confirm whether patients with synchronous peritoneal and hepatic metastases are offered survival benefit from complex surgical intervention (CRS plus HIPEC combined with hepatic resection or RFA).
Collapse
Affiliation(s)
| | | | | | - Alexandros Iliadis
- Laboratory of Diagnostic Histopathology, Mitropoleos 115, Thessaloniki 54622, Greece
| | - Anastasia Fotiadou
- Laboratory of Diagnostic Histopathology, Mitropoleos 115, Thessaloniki 54622, Greece
| |
Collapse
|
33
|
Dreikhausen L, Klupsch A, Wiest I, Xiao Q, Schulte N, Betge J, Boch T, Brochhausen C, Gaiser T, Hofheinz RD, Ebert M, Zhan T. Clinical impact of panel gene sequencing on therapy of advanced cancers of the digestive system: a retrospective, single center study. BMC Cancer 2024; 24:526. [PMID: 38664720 PMCID: PMC11046933 DOI: 10.1186/s12885-024-12261-2] [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: 11/27/2023] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Panel gene sequencing is an established diagnostic tool for precision oncology of solid tumors, but its utility for the treatment of cancers of the digestive system in clinical routine is less well documented. METHODS We retrospectively identified patients with advanced or metastatic gastrointestinal, pancreaticobiliary or hepatic cancers who received panel gene sequencing at a tertiary university hospital from 2015 to 2022. For these cases, we determined the spectrum of genetic alterations, clinicopathological parameters and treatment courses. Assessment of actionability of genetic alterations was based on the OncoKB database, cancer-specific ESMO treatment guidelines, and recommendations of the local molecular tumor board. RESULTS In total, 155 patients received panel gene sequencing using either the Oncomine Focus (62 cases), Comprehensive (91 cases) or Childhood Cancer Research Assay (2 cases). The mean age of patients was 61 years (range 24-90) and 37% were female. Most patients suffered from either colorectal cancer (53%) or cholangiocellular carcinoma (19%). 327 genetic alterations were discovered in 123 tumor samples, with an average number of 2.1 alterations per tumor. The most frequently altered genes were TP53, KRAS and PIK3CA. Actionable gene alterations were detected in 13.5-56.8% of tumors, according to ESMO guidelines or the OncoKB database, respectively. Thirteen patients were treated with targeted therapies based on identified molecular alterations, with a median progression-free survival of 8.8 months. CONCLUSIONS Actionable genetic alterations are frequently detected by panel gene sequencing in patients with advanced cancers of the digestive tract, providing clinical benefit in selected cases. However, for the majority of identified actionable alterations, sufficient clinical evidence for targeted treatments is still lacking.
Collapse
Affiliation(s)
- Lena Dreikhausen
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anna Klupsch
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Isabella Wiest
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Qiyun Xiao
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Nadine Schulte
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Johannes Betge
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center, Heidelberg, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Medical Faculty Mannheim, DKFZ-Hector Cancer Institute, Heidelberg University, Mannheim, Germany
| | - Tobias Boch
- Department of Medicine III, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Medical Faculty Mannheim, DKFZ-Hector Cancer Institute, Heidelberg University, Mannheim, Germany
| | - Christoph Brochhausen
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Timo Gaiser
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ralf-Dieter Hofheinz
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Medicine III, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias Ebert
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Medical Faculty Mannheim, DKFZ-Hector Cancer Institute, Heidelberg University, Mannheim, Germany
| | - Tianzuo Zhan
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Medical Faculty Mannheim, DKFZ-Hector Cancer Institute, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
34
|
Brozos-Vázquez E, Toledano-Fonseca M, Costa-Fraga N, García-Ortiz MV, Díaz-Lagares Á, Rodríguez-Ariza A, Aranda E, López-López R. Pancreatic cancer biomarkers: A pathway to advance in personalized treatment selection. Cancer Treat Rev 2024; 125:102719. [PMID: 38490088 DOI: 10.1016/j.ctrv.2024.102719] [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: 01/08/2024] [Revised: 03/08/2024] [Accepted: 03/10/2024] [Indexed: 03/17/2024]
Abstract
Pancreatic cancer is one of the tumors with the worst prognosis, and unlike other cancers, few advances have been made in recent years. The only curative option is surgery, but only 15-20% of patients are candidates, with a high risk of relapse. In advanced pancreatic cancer there are few first-line treatment options and no validated biomarkers for better treatment selection. The development of targeted therapies in pancreatic cancer is increasingly feasible due to tumor-agnostic treatments, such as PARP inhibitors in patients with BRCA1, BRCA2 or PALB2 alterations or immunotherapies in patients with high microsatellite instability/tumor mutational burden. In addition, other therapeutic molecules have been developed for patients with KRAS G12C mutation or fusions in NTRK or NRG1. Consequently, there has been a growing interest in biomarkers that may help guide targeted therapy in pancreatic cancer. Therefore, this review aims to offer an updated perspective on biomarkers with therapeutic potential in pancreatic cancer.
Collapse
Affiliation(s)
- Elena Brozos-Vázquez
- Medical Oncology Department, University Hospital of A Coruña (CHUAC), A Coruña, Spain
| | - Marta Toledano-Fonseca
- Cancer Network Biomedical Research Center (CIBERONC), Madrid, Spain; Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
| | - Nicolás Costa-Fraga
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (ONCOMET); Clinical University Hospital & Health Research Institute of Santiago de Compostela. CIBERONC; University of Santiago de Compostela, Santiago de Compostela, Spain
| | - María Victoria García-Ortiz
- Cancer Network Biomedical Research Center (CIBERONC), Madrid, Spain; Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
| | - Ángel Díaz-Lagares
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (ONCOMET); Clinical University Hospital & Health Research Institute of Santiago de Compostela. CIBERONC; Department of Clinical Analysis, University Hospital Complex of Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | - Antonio Rodríguez-Ariza
- Cancer Network Biomedical Research Center (CIBERONC), Madrid, Spain; Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Medical Oncology Department, Reina Sofía University Hospital, Córdoba, Spain.
| | - Enrique Aranda
- Cancer Network Biomedical Research Center (CIBERONC), Madrid, Spain; Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Medical Oncology Department, Reina Sofía University Hospital, Córdoba, Spain; Department of Medicine, Faculty of Medicine, University of Córdoba, Córdoba, Spain
| | - Rafael López-López
- Clinical University Hospital & Health Research Institute of Santiago de Compostela. CIBERONC; Medical Oncology Department & Translational Medical Oncology Group-ONCOMET, Spain; Oncology at Santiago de Compostela School of Medicine, Spain
| |
Collapse
|
35
|
Knipper K, Hussein Y, Simon AG, Fretter C, Damanakis AI, Zhao Y, Bruns CJ, Schmidt T, Popp FC, Quaas A, Lyu SI. Upregulation of the histone γ-H2AX correlates with worse patient survival and basal-like subtype in pancreatic ductal adenocarcinoma. J Cancer Res Clin Oncol 2024; 150:137. [PMID: 38502354 PMCID: PMC10950965 DOI: 10.1007/s00432-024-05681-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: 02/03/2024] [Accepted: 03/03/2024] [Indexed: 03/21/2024]
Abstract
PURPOSE Patients with pancreatic ductal adenocarcinoma (PDAC) have yet to experience significant benefits from targeted therapy. Olaparib is currently the only active substance in BRCA-mutated PDACs that successfully influences the DNA repair of carcinoma cells. H2AX belongs to the histone family and is known as a part of the DNA repair system. The inhibition of γ-H2AX could lead to the inhibition of mitotically active tumor cells. Therefore, we aimed to evaluate the predictive value of the γ-H2AX in patients with PDAC. METHODS All included patients (n = 311) received a pancreatic resection with curative intention in one of our PANCALYZE study centers. Subsequently, they were enrolled in a standardized follow-up protocol. Immunohistochemical stainings for γ-H2AX were conducted on tissue microarrays. RESULTS Patients exhibiting high levels of γ-H2AX expression experience more frequent R1 resections, indicating advanced tumor stages in this subgroup. Additionally, patients with high γ-H2AX expression demonstrated significantly poorer survival compared to those with low expression (median OS: 15 vs. 25 months, p < 0.001). In multivariate analyses, high γ-H2AX expression could be identified as an independent risk factor for worse patient survival. Moreover, high γ-H2AX expression could be more frequently observed in the more aggressive basal-like subtype. CONCLUSION γ-H2AX can be characterized as a predictive biomarker for poorer patient survival. Consequently, upcoming clinical trials focused on the efficacy of targeted therapies influencing the DNA repair system and radiotherapy should evaluate γ-H2AX as a potential biomarker for therapy response. Furthermore, γ-H2AX may serve as a viable target for treatment in the future.
Collapse
Affiliation(s)
- Karl Knipper
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany.
| | - Yussra Hussein
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Adrian Georg Simon
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Caroline Fretter
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Alexander I Damanakis
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Yue Zhao
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Christiane J Bruns
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Thomas Schmidt
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Felix C Popp
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Su Ir Lyu
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| |
Collapse
|
36
|
Ikezawa K, Urabe M, Kai Y, Takada R, Akita H, Nagata S, Ohkawa K. Comprehensive review of pancreatic acinar cell carcinoma: epidemiology, diagnosis, molecular features and treatment. Jpn J Clin Oncol 2024; 54:271-281. [PMID: 38109477 PMCID: PMC10925851 DOI: 10.1093/jjco/hyad176] [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: 09/26/2023] [Accepted: 11/29/2023] [Indexed: 12/20/2023] Open
Abstract
Pancreatic acinar cell carcinoma is a rare form (0.2-4.3%) of pancreatic neoplasm with unique clinical and molecular characteristics, which largely differ from pancreatic ductal adenocarcinoma. Pancreatic acinar cell carcinoma occurs more frequently in males and can occur in children. Serum lipase is elevated in 24-58% of patients with pancreatic acinar cell carcinoma. Pancreatic acinar cell carcinomas tend to be large at diagnosis (median tumour size: ~5 cm) and are frequently located in the pancreas head. Radiologically, pancreatic acinar cell carcinoma generally exhibits a solid appearance; however, necrosis, cystic changes and intratumoral haemorrhage can occur in larger lesions. Immunostaining is essential for the definitive diagnosis of pancreatic acinar cell carcinoma. Compared with pancreatic ductal adenocarcinoma, pancreatic acinar cell carcinoma has a more favourable prognosis. Although radical surgery is recommended for patients with pancreatic acinar cell carcinoma who do not have distant metastases, the recurrence rate is high. The effectiveness of adjuvant therapy for pancreatic acinar cell carcinoma is unclear. The response to FOLFIRINOX is generally favourable, and some patients achieve a complete response. Pancreatic acinar cell carcinoma has a different genomic profile compared with pancreatic ductal adenocarcinoma. Although genomic analyses have shown that pancreatic acinar cell carcinoma rarely has KRAS, TP53 and CDKN2A mutations, it has a higher prevalence of homologous recombination-related genes, including BRCA1/2 and ATM, than pancreatic ductal adenocarcinoma, suggesting high sensitivity to platinum-containing regimens and PARP inhibitors. Targeted therapies for genomic alternations are beneficial. Therefore, genetic testing is important for patients with pancreatic acinar cell carcinoma to choose the optimal therapeutic strategy.
Collapse
Affiliation(s)
- Kenji Ikezawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Makiko Urabe
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Yugo Kai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Ryoji Takada
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Hirofumi Akita
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Shigenori Nagata
- Department of Diagnostic Pathology and Cytology, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuyoshi Ohkawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
37
|
Anbil S, Reiss KA. Targeting BRCA and PALB2 in Pancreatic Cancer. Curr Treat Options Oncol 2024; 25:346-363. [PMID: 38311708 DOI: 10.1007/s11864-023-01174-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2023] [Indexed: 02/06/2024]
Abstract
OPINION STATEMENT An important subgroup of pancreatic ductal adenocarcinomas (PDACs) harbor pathogenic variants in BRCA1, BRCA2, or PALB2. These tumors are exquisitely sensitive to platinum-based chemotherapy and patients may experience deep and durable responses to this treatment. PARP inhibitors offer potential respite from the cumulative toxicities of chemotherapy as they significantly extend progression-free survival compared to a chemotherapy holiday. Given the lack of proven survival benefit, the decision to use a maintenance PARP inhibitor rather than continue chemotherapy should be individualized. Interestingly, in both published clinical trials of maintenance PARP inhibitors, there is a striking range of interpatient benefit: Even in the platinum-sensitive setting, roughly 25% of tumors appear to be PARP inhibitor refractory (progressive disease within 2 months of starting treatment), 50% sustain moderate benefit (up to 2 years), and 25% are hyper-responsive (more than 2 years of benefit). This finding highlights the need to refine our understanding of which patients will respond to maintenance PARP inhibitors, both by being able to identify biallelic loss and by deepening our knowledge of resistance mechanisms and who develops them. Recent data supports that reversion mutations are common in PARP inhibitor refractory patients, but we have little understanding of the mechanisms that drive delayed resistance and long-term responses. Identifying which patients are more prone to certain mechanisms of resistance and tackling them with specific treatment strategies are areas of active investigation. Additionally, given that PARP inhibitors have limited overall efficacy for most patients, upfront combination strategies are an important future strategy.
Collapse
Affiliation(s)
- Sriram Anbil
- Abramson Cancer Center, 10th Floor Perelman Center South, The University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19121, USA
| | - Kim A Reiss
- Abramson Cancer Center, 10th Floor Perelman Center South, The University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19121, USA.
| |
Collapse
|
38
|
Reni M, Peretti U, Macchini M, Orsi G, Militello A, Briccolani A, Falconi M, Cascinu S. Cyclophosphamide maintenance to extend combination chemotherapy-free interval in metastatic pancreatic ductal adenocarcinoma. Dig Liver Dis 2024; 56:509-513. [PMID: 37586911 DOI: 10.1016/j.dld.2023.07.033] [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: 05/22/2023] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Administering chemotherapy until progression to metastatic pancreatic ductal adenocarcinoma (PDAC) patients lacks of supporting evidence and causes cumulative toxicity. We explored the role of cyclophosphamide as maintenance therapy. METHODS PDAC germline BRCA1-2 wild-type patients who were progression-free after ≥6 months of any regimen and line of chemotherapy and received maintenance cyclophosphamide (mCTX) (50 mg/day), were included in the analysis. RESULTS 42 patients were included in the analysis. Thirty-nine patients had progression of disease. Median PFS was 3.5 (range 1.0-31+) months. PFS rates at 6 and 12 months were 26.2% and 11.9%. At a median follow-up of 20.0 months (range 12.1-31.0 months), 20 patients died and 22 are alive. Median OS was 20.0 months (range 2.2-31.0+). OS at 6 and 12 months was 97.6% (95%CI: 93.4-100), and 73.8% (95% CI: 61.1-86.5), respectively. Only 2 patients receiving mCTX had Grade 3 toxicity. CONCLUSIONS mCTX therapy yielded promising PFS and OS outcome in PDAC patients who were progression-free after induction chemotherapy, with unremarkable toxicity. Accordingly, this approach warrants further investigation.
Collapse
Affiliation(s)
- Michele Reni
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| | - Umberto Peretti
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Marina Macchini
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Giulia Orsi
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Annamaria Militello
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Assunta Briccolani
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Falconi
- Vita-Salute San Raffaele University, Milan, Italy; Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Cascinu
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
39
|
Gyawali B, Booth CM. Treatment of metastatic pancreatic cancer: 25 years of innovation with little progress for patients. Lancet Oncol 2024; 25:167-170. [PMID: 38301687 DOI: 10.1016/s1470-2045(23)00516-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 02/03/2024]
Affiliation(s)
- Bishal Gyawali
- Division of Cancer Care and Epidemiology, Queen's University Cancer Research Institute, Kingston, ON K7L 3N6, Canada; Department of Oncology and Department of Public Health Sciences, Queen's University, Kingston, ON, Canada.
| | - Christopher M Booth
- Division of Cancer Care and Epidemiology, Queen's University Cancer Research Institute, Kingston, ON K7L 3N6, Canada; Department of Oncology and Department of Public Health Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
40
|
LaRose M, Manji GA, Bates SE. Beyond BRCA: Diagnosis and management of homologous recombination repair deficient pancreatic cancer. Semin Oncol 2024; 51:36-44. [PMID: 38171988 DOI: 10.1053/j.seminoncol.2023.11.001] [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: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 01/05/2024]
Abstract
Approximately 4%-7% of patients diagnosed with pancreatic adenocarcinoma (PDAC) are found to harbor deleterious germline mutations in BRCA1 and/or BRCA2. Loss of function of BRCA1 and/or BRCA2 results in deficiency in homologous recombination repair (HRR), a critical DNA repair pathway, and confers sensitivity to certain DNA damaging agents, including platinum chemotherapy and PARP inhibitors. The PARP inhibitor olaparib is food and drug administration (FDA) approved for use in pancreatic cancer based on the POLO trial, which found that maintenance olaparib significantly prolonged progression free survival compared to placebo among patients with germline BRCA1 or BRCA2 mutations and metastatic PDAC that had not progressed following frontline platinum-based chemotherapy. Recently, there has been considerable interest in identifying patients without BRCA inactivation whose tumors also exhibit properties of HRR deficiency and thus may be susceptible to therapies with proven benefit in cancers harboring BRCA mutations. Here, we discuss methods for identification of HRR-deficiency and review the management of HRR-deficient cancers with a focus on HRR-deficient PDAC.
Collapse
Affiliation(s)
- Meredith LaRose
- Columbia University Irving Medical Center, New York NY, USA.
| | - Gulam A Manji
- Columbia University Irving Medical Center, New York NY, USA
| | - Susan E Bates
- Columbia University Irving Medical Center, New York NY, USA
| |
Collapse
|
41
|
Khamidullina AI, Abramenko YE, Bruter AV, Tatarskiy VV. Key Proteins of Replication Stress Response and Cell Cycle Control as Cancer Therapy Targets. Int J Mol Sci 2024; 25:1263. [PMID: 38279263 PMCID: PMC10816012 DOI: 10.3390/ijms25021263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/14/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024] Open
Abstract
Replication stress (RS) is a characteristic state of cancer cells as they tend to exchange precision of replication for fast proliferation and increased genomic instability. To overcome the consequences of improper replication control, malignant cells frequently inactivate parts of their DNA damage response (DDR) pathways (the ATM-CHK2-p53 pathway), while relying on other pathways which help to maintain replication fork stability (ATR-CHK1). This creates a dependency on the remaining DDR pathways, vulnerability to further destabilization of replication and synthetic lethality of DDR inhibitors with common oncogenic alterations such as mutations of TP53, RB1, ATM, amplifications of MYC, CCNE1 and others. The response to RS is normally limited by coordination of cell cycle, transcription and replication. Inhibition of WEE1 and PKMYT1 kinases, which prevent unscheduled mitosis entry, leads to fragility of under-replicated sites. Recent evidence also shows that inhibition of Cyclin-dependent kinases (CDKs), such as CDK4/6, CDK2, CDK8/19 and CDK12/13 can contribute to RS through disruption of DNA repair and replication control. Here, we review the main causes of RS in cancers as well as main therapeutic targets-ATR, CHK1, PARP and their inhibitors.
Collapse
Affiliation(s)
- Alvina I. Khamidullina
- Laboratory of Molecular Oncobiology, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia; (A.I.K.); (Y.E.A.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia
| | - Yaroslav E. Abramenko
- Laboratory of Molecular Oncobiology, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia; (A.I.K.); (Y.E.A.)
| | - Alexandra V. Bruter
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia
| | - Victor V. Tatarskiy
- Laboratory of Molecular Oncobiology, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia; (A.I.K.); (Y.E.A.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia
| |
Collapse
|
42
|
Guan X, Li Y, Guan X, Fan L, Ying J. XYA-2: a marine-derived compound targeting apoptosis and multiple signaling pathways in pancreatic cancer. PeerJ 2024; 12:e16805. [PMID: 38250721 PMCID: PMC10798151 DOI: 10.7717/peerj.16805] [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: 10/20/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Background Pancreatic cancer is a highly aggressive and fatal disease with limited treatment options and poor prognosis for patients. This study aimed to investigate the impact of XYA-2 {N-(3,7-dimethyl-2,6-octadienyl)-2-aza-2-deoxychaetoviridin A}, a nitrogenated azaphilon previously reported from a deep-sea-derived fungus on the progression of pancreatic cancer cells. Methods The inhibitory effects of XYA-2 on cell proliferation, clonogenic potential, cell cycle progression, apoptosis, migration, and invasion were assessed using various assays. The CCK-8 assay, clone formation assay, flow cytometry assay, wound healing assay, and transwell assay were employed to evaluate cell proliferation, clonogenic potential, cell cycle progression, apoptosis, migration, and invasion, respectively. Moreover, we employed RNA-seq and bioinformatics analyses to uncover the underlying mechanism by which XYA-2 influences pancreatic cancer cells. The revealed mechanism was subsequently validated through qRT-PCR. Results Our results demonstrated that XYA-2 dose-dependently inhibited the proliferation of pancreatic cancer cells and induced cell cycle arrest and apoptosis. Additionally, XYA-2 exerted a significant inhibitory effect on the invasion and migration of cancer cells. Moreover, XYA-2 was found to regulate the expression of genes involved in multiple cancer-related pathways based on our RNA-seq and bioinformatics analysis. Conclusion These findings highlight the potential of XYA-2 as a promising therapeutic option for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Xiaoqing Guan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Zhejiang, China
| | - Yun Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
| | - Xiaodan Guan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
| | - Linfei Fan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
| | - Jieer Ying
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Zhejiang, China
| |
Collapse
|
43
|
Buckley CW, O’Reilly EM. Next-generation therapies for pancreatic cancer. Expert Rev Gastroenterol Hepatol 2024; 18:55-72. [PMID: 38415709 PMCID: PMC10960610 DOI: 10.1080/17474124.2024.2322648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/20/2024] [Indexed: 02/29/2024]
Abstract
INTRODUCTION Pancreas ductal adenocarcinoma (PDAC) is a frequently lethal malignancy that poses unique therapeutic challenges. The current mainstay of therapy for metastatic PDAC (mPDAC) is cytotoxic chemotherapy. NALIRIFOX (liposomal irinotecan, fluorouracil, leucovorin, oxaliplatin) is an emerging standard of care in the metastatic setting. An evolving understanding of PDAC pathogenesis is driving a shift toward targeted therapy. Olaparib, a poly-ADP-ribose polymerase (PARP) inhibitor, has regulatory approval for maintenance therapy in BRCA-mutated mPDAC along with other targeted agents receiving disease-agnostic approvals including for PDAC with rare fusions and mismatch repair deficiency. Ongoing research continues to identify and evaluate an expanding array of targeted therapies for PDAC. AREAS COVERED This review provides a brief overview of standard therapies for PDAC and an emphasis on current and emerging targeted therapies. EXPERT OPINION There is notable potential for targeted therapies for KRAS-mutated PDAC with opportunity for meaningful benefit for a sizable portion of patients with this disease. Further, emerging approaches are focused on novel immune, tumor microenvironment, and synthetic lethality strategies.
Collapse
Affiliation(s)
- Conor W. Buckley
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Eileen M. O’Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
- Weill Cornell Medicine, New York, USA
| |
Collapse
|
44
|
Hu ZI, O'Reilly EM. Therapeutic developments in pancreatic cancer. Nat Rev Gastroenterol Hepatol 2024; 21:7-24. [PMID: 37798442 DOI: 10.1038/s41575-023-00840-w] [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] [Accepted: 08/15/2023] [Indexed: 10/07/2023]
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
|
45
|
Tsang ES, Gallinger S. Deciphering the Pathways to PARP Sensitivity in Pancreatic Cancer. Clin Cancer Res 2023; 29:5005-5007. [PMID: 37787975 DOI: 10.1158/1078-0432.ccr-23-2260] [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: 08/15/2023] [Revised: 09/01/2023] [Accepted: 09/15/2023] [Indexed: 10/04/2023]
Abstract
A recent article analyzed paired cell-free DNA among patients with platinum-sensitive BRCA- or PALB2-mutated pancreatic cancer who received maintenance olaparib. Reversion mutations were linked with worse outcomes. These types of paired correlative studies are needed to improve our understanding of drug resistance and guide future clinical trials. See related article by Brown et al., p. 5207.
Collapse
Affiliation(s)
- Erica S Tsang
- Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
46
|
Del Chiaro M, Sugawara T, Karam SD, Messersmith WA. Advances in the management of pancreatic cancer. BMJ 2023; 383:e073995. [PMID: 38164628 DOI: 10.1136/bmj-2022-073995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Pancreatic cancer remains among the malignancies with the worst outcomes. Survival has been improving, but at a slower rate than other cancers. Multimodal treatment, including chemotherapy, surgical resection, and radiotherapy, has been under investigation for many years. Because of the anatomical characteristics of the pancreas, more emphasis on treatment selection has been placed on local extension into major vessels. Recently, the development of more effective treatment regimens has opened up new treatment strategies, but urgent research questions have also become apparent. This review outlines the current management of pancreatic cancer, and the recent advances in its treatment. The review discusses future treatment pathways aimed at integrating novel findings of translational and clinical research.
Collapse
Affiliation(s)
- Marco Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, USA
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Toshitaka Sugawara
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sana D Karam
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Wells A Messersmith
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
47
|
Hilmi M, Delaye M, Muzzolini M, Nicolle R, Cros J, Hammel P, Cardot-Ruffino V, Neuzillet C. The immunological landscape in pancreatic ductal adenocarcinoma and overcoming resistance to immunotherapy. Lancet Gastroenterol Hepatol 2023; 8:1129-1142. [PMID: 37866368 DOI: 10.1016/s2468-1253(23)00207-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 10/24/2023]
Abstract
Pancreatic ductal adenocarcinoma is associated with a poor prognosis and there are few treatment options. The development of immunotherapy in pancreatic ductal adenocarcinoma has been difficult, and immune checkpoint inhibitors are only effective in a very small subset of patients. Most obstacles for treatment have been related to intertumoural and intratumoural heterogeneity, the composition of tumour stroma, and crosstalk with cancer cells. Improved molecular characterisation of pancreatic ductal adenocarcinoma and a better understanding of its microenvironment have paved the way for novel immunotherapy strategies, including the identification of predictive biomarkers, the development of rational combinations to optimise effectiveness, and the targeting of new mechanisms. Future immunotherapy strategies should consider individual characteristics to move beyond the traditional immune targets and circumvent the resistance to therapies that have been developed so far.
Collapse
Affiliation(s)
- Marc Hilmi
- Gastrointestinal Oncology, Medical Oncology Department, Institut Curie, Université Versailles Saint-Quentin-Université Paris-Saclay, Saint-Cloud, France; Molecular Oncology, PSL Research University, CNRS, UMR 144, Institut Curie, Paris, France
| | - Matthieu Delaye
- Gastrointestinal Oncology, Medical Oncology Department, Institut Curie, Université Versailles Saint-Quentin-Université Paris-Saclay, Saint-Cloud, France; Molecular Oncology, PSL Research University, CNRS, UMR 144, Institut Curie, Paris, France
| | - Milena Muzzolini
- Digestive Surgery Department, Ambroise Paré Hospital, APHP, Université Versailles Saint-Quentin-Université Paris-Saclay, Boulogne Billancourt, France
| | - Rémy Nicolle
- Université Paris Cité, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, Paris, France
| | - Jérôme Cros
- Université Paris Cité, Pathology Department, Beaujon Hospital, FHU MOSAIC, AP-HP, Clichy, France
| | - Pascal Hammel
- Université Paris-Saclay, Department of Digestive and Medical Oncology, Paul-Brousse Hospital (APHP Sud), Villejuif, France
| | | | - Cindy Neuzillet
- Gastrointestinal Oncology, Medical Oncology Department, Institut Curie, Université Versailles Saint-Quentin-Université Paris-Saclay, Saint-Cloud, France; Molecular Oncology, PSL Research University, CNRS, UMR 144, Institut Curie, Paris, France.
| |
Collapse
|
48
|
Tsimberidou AM, Kahle M, Vo HH, Baysal MA, Johnson A, Meric-Bernstam F. Molecular tumour boards - current and future considerations for precision oncology. Nat Rev Clin Oncol 2023; 20:843-863. [PMID: 37845306 DOI: 10.1038/s41571-023-00824-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2023] [Indexed: 10/18/2023]
Abstract
Over the past 15 years, rapid progress has been made in developmental therapeutics, especially regarding the use of matched targeted therapies against specific oncogenic molecular alterations across cancer types. Molecular tumour boards (MTBs) are panels of expert physicians, scientists, health-care providers and patient advocates who review and interpret molecular-profiling results for individual patients with cancer and match each patient to available therapies, which can include investigational drugs. Interpretation of the molecular alterations found in each patient is a complicated task that requires an understanding of their contextual functional effects and their correlations with sensitivity or resistance to specific treatments. The criteria for determining the actionability of molecular alterations and selecting matched treatments are constantly evolving. Therefore, MTBs have an increasingly necessary role in optimizing the allocation of biomarker-directed therapies and the implementation of precision oncology. Ultimately, increased MTB availability, accessibility and performance are likely to improve patient care. The challenges faced by MTBs are increasing, owing to the plethora of identifiable molecular alterations and immune markers in tumours of individual patients and their evolving clinical significance as more and more data on patient outcomes and results from clinical trials become available. Beyond next-generation sequencing, broader biomarker analyses can provide useful information. However, greater funding, resources and expertise are needed to ensure the sustainability of MTBs and expand their outreach to underserved populations. Harmonization between practice and policy will be required to optimally implement precision oncology. Herein, we discuss the evolving role of MTBs and current and future considerations for their use in precision oncology.
Collapse
Affiliation(s)
- Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Michael Kahle
- Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Henry Hiep Vo
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mehmet A Baysal
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amber Johnson
- Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
49
|
Zheng S, Tian Q, Yuan Y, Sun S, Li T, Xia R, He R, Luo Y, Lin Q, Fu Z, Zhou Y, Chen R, Hu C. Extracellular vesicle-packaged circBIRC6 from cancer-associated fibroblasts induce platinum resistance via SUMOylation modulation in pancreatic cancer. J Exp Clin Cancer Res 2023; 42:324. [PMID: 38012734 PMCID: PMC10683239 DOI: 10.1186/s13046-023-02854-3] [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: 06/29/2023] [Accepted: 10/07/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) play pivotal roles in chemoresistance of pancreatic ductal adenocarcinoma (PDAC). However, the underlying mechanisms are poorly understood. Revealing the cross-talk network between tumor stroma and pancreatic cancer and developing effective strategies against oxaliplatin resistance are highly desired in the clinic. METHODS High-throughput sequence was used to screened the key circRNAs transmitted by extracellular vesicles (EVs) from CAFs to pancreatic cancer cells. The associations between EV-packaged circBIRC6 and chemotherapy responsiveness were validated in a cohort of 82 cases of advanced PDAC patients. Then, the effects of EV-packaged circBIRC6 on CAF-induced oxaliplatin resistance were investigated by flow cytometry, colony formation, viability of pancreatic cancer organoids in vitro and by xenograft models in vivo. RNA pulldown, RNA immunoprecipitation, and sites mutation assays were used to reveal the underlying mechanism. RESULTS We identified a circRNA, circBIRC6, is significantly upregulated in CAF-derived EVs and is positively associated with oxaliplatin-based chemoresistance. In vitro and in vivo functional assays showed that CAF-derived EV-packaged circBIRC6 enhance oxaliplatin resistance of pancreatic cancer cells and organoids via regulating the non-homologous end joining (NHEJ) dependent DNA repair. Mechanistically, circBIRC6 directly binds with XRCC4 and enhanced the interaction of XRCC4 with SUMO1 at the lysine 115 residue, which facilitated XRCC4 chromatin localization. XRCC4K115R mutation dramatically abrogated the EV-packaged circBIRC6 induced effect. Moreover, combination of antisense oligonucleotide inhibitors against circBIRC6 with Olaparib dramatically suppressed chemoresistance in patient-derived xenograft models. CONCLUSIONS Our study revealed that EV-packaged circBIRC6 confer oxaliplatin resistance in PDAC by mediating SUMOylation of XRCC4, introducing a promising predictive and therapeutic target for PDAC on oxaliplatin resistance.
Collapse
Affiliation(s)
- Shangyou Zheng
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Qing Tian
- School of medicine, South China University of Technology, Guangzhou, 510006, Guangdong Province, China
| | - Yuan Yuan
- Guangdong cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Shuxin Sun
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Tingting Li
- School of medicine, South China University of Technology, Guangzhou, 510006, Guangdong Province, China
| | - Renpeng Xia
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Rihua He
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China
- Shantou University Medical College, Shantou, 515041, Guangdong province, China
| | - Yuming Luo
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Qing Lin
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Zhiqiang Fu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Yu Zhou
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China.
| | - Rufu Chen
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China.
- School of medicine, South China University of Technology, Guangzhou, 510006, Guangdong Province, China.
- Guangdong cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China.
| | - Chonghui Hu
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
50
|
Booth CM, Eisenhauer EA, Gyawali B, Tannock IF. Progression-Free Survival Should Not Be Used as a Primary End Point for Registration of Anticancer Drugs. J Clin Oncol 2023; 41:4968-4972. [PMID: 37733981 DOI: 10.1200/jco.23.01423] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 09/23/2023] Open
Affiliation(s)
- Christopher M Booth
- Division of Cancer Care and Epidemiology, Queen's University Cancer Research Institute, Kingston, Canada
- Department of Oncology, Queen's University, Kingston, Canada
- Department of Public Health Sciences, Queen's University, Kingston, Canada
| | | | - Bishal Gyawali
- Division of Cancer Care and Epidemiology, Queen's University Cancer Research Institute, Kingston, Canada
- Department of Oncology, Queen's University, Kingston, Canada
- Department of Public Health Sciences, Queen's University, Kingston, Canada
| | - Ian F Tannock
- Division of Medical Oncology, Princess Margaret Cancer Centre and University of Toronto, Toronto, Canada
| |
Collapse
|