1
|
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
|
2
|
Salu P, Reindl KM. Advancements in Preclinical Models of Pancreatic Cancer. Pancreas 2024; 53:e205-e220. [PMID: 38206758 PMCID: PMC10842038 DOI: 10.1097/mpa.0000000000002277] [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] [Indexed: 01/13/2024]
Abstract
ABSTRACT Pancreatic cancer remains one of the deadliest of all cancer types with a 5-year overall survival rate of just 12%. Preclinical models available for understanding the disease pathophysiology have evolved significantly in recent years. Traditionally, commercially available 2-dimensional cell lines were developed to investigate mechanisms underlying tumorigenesis, metastasis, and drug resistance. However, these cells grow as monolayer cultures that lack heterogeneity and do not effectively represent tumor biology. Developing patient-derived xenografts and genetically engineered mouse models led to increased cellular heterogeneity, molecular diversity, and tissues that histologically represent the original patient tumors. However, these models are relatively expensive and very timing consuming. More recently, the advancement of fast and inexpensive in vitro models that better mimic disease conditions in vivo are on the rise. Three-dimensional cultures like organoids and spheroids have gained popularity and are considered to recapitulate complex disease characteristics. In addition, computational genomics, transcriptomics, and metabolomic models are being developed to simulate pancreatic cancer progression and predict better treatment strategies. Herein, we review the challenges associated with pancreatic cancer research and available analytical models. We suggest that an integrated approach toward using these models may allow for developing new strategies for pancreatic cancer precision medicine.
Collapse
Affiliation(s)
- Philip Salu
- From the Department of Biological Sciences, North Dakota State University, Fargo, ND
| | | |
Collapse
|
3
|
Hartupee C, Nagalo BM, Chabu CY, Tesfay MZ, Coleman-Barnett J, West JT, Moaven O. Pancreatic cancer tumor microenvironment is a major therapeutic barrier and target. Front Immunol 2024; 15:1287459. [PMID: 38361931 PMCID: PMC10867137 DOI: 10.3389/fimmu.2024.1287459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/04/2024] [Indexed: 02/17/2024] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is projected to become the 2nd leading cause of cancer-related deaths in the United States. Limitations in early detection and treatment barriers contribute to the lack of substantial success in the treatment of this challenging-to-treat malignancy. Desmoplasia is the hallmark of PDAC microenvironment that creates a physical and immunologic barrier. Stromal support cells and immunomodulatory cells face aberrant signaling by pancreatic cancer cells that shifts the complex balance of proper repair mechanisms into a state of dysregulation. The product of this dysregulation is the desmoplastic environment that encases the malignant cells leading to a dense, hypoxic environment that promotes further tumorigenesis, provides innate systemic resistance, and suppresses anti-tumor immune invasion. This desmoplastic environment combined with the immunoregulatory events that allow it to persist serve as the primary focus of this review. The physical barrier and immune counterbalance in the tumor microenvironment (TME) make PDAC an immunologically cold tumor. To convert PDAC into an immunologically hot tumor, tumor microenvironment could be considered alongside the tumor cells. We discuss the complex network of microenvironment molecular and cellular composition and explore how they can be targeted to overcome immuno-therapeutic challenges.
Collapse
Affiliation(s)
- Conner Hartupee
- Division of Surgical Oncology, Department of Surgery, Louisiana State University (LSU) Health, New Orleans, LA, United States
| | - Bolni Marius Nagalo
- Department of Pathology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, United States
- The Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, United States
| | - Chiswili Y. Chabu
- Division of Biological Sciences, University of Missouri, Columbia, MO, United States
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, United States
- Siteman Cancer Center, Washington University, St. Louis, MO, United States
| | - Mulu Z. Tesfay
- Department of Pathology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, United States
| | - Joycelynn Coleman-Barnett
- Division of Surgical Oncology, Department of Surgery, Louisiana State University (LSU) Health, New Orleans, LA, United States
- Department of Interdisciplinary Oncology, Louisiana Cancer Research Center, Louisiana State University (LSU) Health, New Orleans, LA, United States
| | - John T. West
- Department of Interdisciplinary Oncology, Louisiana Cancer Research Center, Louisiana State University (LSU) Health, New Orleans, LA, United States
| | - Omeed Moaven
- Division of Surgical Oncology, Department of Surgery, Louisiana State University (LSU) Health, New Orleans, LA, United States
- Department of Interdisciplinary Oncology, Louisiana Cancer Research Center, Louisiana State University (LSU) Health, New Orleans, LA, United States
- Louisiana State University - Louisiana Children's Medical Center (LSU - LCMC) Cancer Center, New Orleans, LA, United States
| |
Collapse
|
4
|
Tang HY, Cao YZ, Zhou YW, Ma YS, Jiang H, Zhang H, Jiang L, Yang QX, Tang XM, Yang C, Liu XY, Liu FX, Liu JB, Fu D, Wang YF, Yu H. The power and the promise of CAR-mediated cell immunotherapy for clinical application in pancreatic cancer. J Adv Res 2024:S2090-1232(24)00027-4. [PMID: 38244773 DOI: 10.1016/j.jare.2024.01.014] [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: 10/18/2023] [Revised: 12/24/2023] [Accepted: 01/11/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Pancreatic cancer, referred to as the "monarch of malignancies," is a neoplastic growth mostly arising from the epithelial cells of the pancreatic duct and acinar cells. This particular neoplasm has a highly unfavorable prognosis due to its marked malignancy, inconspicuous initial manifestation, challenging early detection, rapid advancement, and limited survival duration. Cellular immunotherapy is the ex vivo culture and expansion of immune effector cells, granting them the capacity to selectively target malignant cells using specialized techniques. Subsequently, these modified cells are reintroduced into the patient's organism with the purpose of eradicating tumor cells and providing therapeutic intervention for cancer. PRESENT SITUATION Presently, the primary cellular therapeutic modalities employed in the treatment of pancreatic cancer encompass CAR T-cell therapy, TCR T-cell therapy, NK-cell therapy, and CAR NK-cell therapy. AIM OF REVIEW This review provides a concise overview of the mechanisms and primary targets associated with various cell therapies. Additionally, we will explore the prospective outlook of cell therapy in the context of treating pancreatic cancer.
Collapse
Affiliation(s)
- Hao-Yu Tang
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China; Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226631, Jiangsu, China; General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, Shanghai, China
| | - Yi-Zhi Cao
- General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, Shanghai, China
| | - Yi-Wei Zhou
- General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, Shanghai, China
| | - Yu-Shui Ma
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, Shanghai, China
| | - Hong Jiang
- Department of Thoracic Surgery, The 905th Hospital of Chinese People's Liberation Army Navy, Shanghai 200050, Shanghai, China
| | - Hui Zhang
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226631, Jiangsu, China
| | - Lin Jiang
- Department of Anesthesiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu 225300, China
| | - Qin-Xin Yang
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China
| | - Xiao-Mei Tang
- General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, Shanghai, China
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xin-Yun Liu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China
| | - Fu-Xing Liu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China
| | - Ji-Bin Liu
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226631, Jiangsu, China.
| | - Da Fu
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226631, Jiangsu, China; General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, Shanghai, China.
| | - Yun-Feng Wang
- Department of General Surgery, Pudong New Area People's Hospital, Shanghai 201299, China.
| | - Hong Yu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China; Department of Pathology, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou 225300, Jiangsu, China.
| |
Collapse
|
5
|
Liu Y, Peng C, Ahad F, Ali Zaidi SA, Muluh TA, Fu Q. Advanced Strategies of CAR-T Cell Therapy in Solid Tumors and Hematological Malignancies. Recent Pat Anticancer Drug Discov 2024; 19:557-572. [PMID: 38213150 DOI: 10.2174/0115748928277331231218115402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/30/2023] [Accepted: 11/10/2023] [Indexed: 01/13/2024]
Abstract
Chimeric antigen receptor T-cells, known as CAR-T cells, represent a promising breakthrough in the realm of adoptive cell therapy. These T-cells are genetically engineered to carry chimeric antigen receptors that specifically target tumors. They have achieved notable success in the treatment of blood-related cancers, breathing new life into this field of medical research. However, numerous obstacles limit chimeric antigen receptors T-cell therapy's efficacy, such as it cannot survive in the body long. It is prone to fatigue and exhaustion, leading to difficult tumor elimination and repeated recurrence, affecting solid tumors and hematological malignancies. The challenges posed by solid tumors, especially in the context of the complex solid-tumor microenvironment, require specific strategies. This review outlines recent advancements in improving chimeric antigen receptors T-cell therapy by focusing on the chimeric antigen receptors protein, modifying T-cells, and optimizing the interaction between T-cells and other components within the tumor microenvironment. This article aims to provide an extensive summary of the latest discoveries regarding CAR-T cell therapy, encompassing its application across various types of human cancers. Moreover, it will delve into the obstacles that have emerged in recent times, offering insights into the challenges faced by this innovative approach. Finally, it highlights novel therapeutic options in treating hematological and solid malignancies with chimeric antigen receptors T-cell therapies.
Collapse
Affiliation(s)
- Yangjie Liu
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan, PRC China
| | - Cao Peng
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan PRC China
| | - Faiza Ahad
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Syed Aqib Ali Zaidi
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Tobias Achu Muluh
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Qiuxia Fu
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan PRC China
| |
Collapse
|
6
|
Leowattana W, Leowattana P, Leowattana T. Systemic treatment for advanced pancreatic cancer. World J Gastrointest Oncol 2023; 15:1691-1705. [PMID: 37969416 PMCID: PMC10631439 DOI: 10.4251/wjgo.v15.i10.1691] [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: 07/11/2023] [Revised: 08/24/2023] [Accepted: 09/22/2023] [Indexed: 10/10/2023] Open
Abstract
Pancreatic cancer is a deadly disease with an extremely poor 5-year survival rate due to treatment resistance and late-stage detection. Despite numerous years of research and pharmaceutical development, these figures have not changed. Treatment options for advanced pancreatic cancer are still limited. This illness is typically detected at a late stage, making curative surgical resection impossible. Chemotherapy is the most commonly utilized technique for treating advanced pancreatic cancer but has poor efficacy. Targeted therapy and immunotherapy have made significant progress in many other cancer types and have been proven to have extremely promising possibilities; these therapies also hold promise for pancreatic cancer. There is an urgent need for research into targeted treatment, immunotherapy, and cancer vaccines. In this review, we emphasize the foundational findings that have fueled the therapeutic strategy for advanced pancreatic cancer. We also address current advancements in targeted therapy, immunotherapy, and cancer vaccines, all of which continue to improve the clinical outcome of advanced pancreatic cancer. We believe that clinical translation of these novel treatments will improve the low survival rate of this deadly disease.
Collapse
Affiliation(s)
- Wattana Leowattana
- Department of Clinical Tropical Medicine, Mahidol University, Rachatawee 10400, Bangkok, Thailand
| | - Pathomthep Leowattana
- Department of Clinical Tropical Medicine, Mahidol University, Rachatawee 10400, Bangkok, Thailand
| | - Tawithep Leowattana
- Department of Medicine, Srinakharinwirot University, Wattana 10110, Bangkok, Thailand
| |
Collapse
|
7
|
Yue NN, Xu HM, Xu J, Zhu MZ, Zhang Y, Tian CM, Nie YQ, Yao J, Liang YJ, Li DF, Wang LS. Therapeutic potential of gene therapy for gastrointestinal diseases: Advancements and future perspectives. Mol Ther Oncolytics 2023; 30:193-215. [PMID: 37663132 PMCID: PMC10471515 DOI: 10.1016/j.omto.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023] Open
Abstract
Advancements in understanding the pathogenesis mechanisms underlying gastrointestinal diseases, encompassing inflammatory bowel disease, gastrointestinal cancer, and gastroesophageal reflux disease, have led to the identification of numerous novel therapeutic targets. These discoveries have opened up exciting possibilities for developing gene therapy strategies to treat gastrointestinal diseases. These strategies include gene replacement, gene enhancement, gene overexpression, gene function blocking, and transgenic somatic cell transplantation. In this review, we introduce the important gene therapy targets and targeted delivery systems within the field of gastroenterology. Furthermore, we provide a comprehensive overview of recent progress in gene therapy related to gastrointestinal disorders and shed light on the application of innovative gene-editing technologies in treating these conditions. These developments are fueling a revolution in the management of gastrointestinal diseases. Ultimately, we discuss the current challenges (particularly regarding safety, oral efficacy, and cost) and explore potential future directions for implementing gene therapy in the clinical settings for gastrointestinal diseases.
Collapse
Affiliation(s)
- Ning-ning Yue
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen 518000, China
| | - Hao-ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
| | - Min-zheng Zhu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, Guangdong 516000, China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, China
| | - Yu-qiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, China
| | - Yu-jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen 518000, China
| | - De-feng Li
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, China
| | - Li-sheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, China
| |
Collapse
|
8
|
Wang J, Cheng X, Jin Y, Xia B, Qin R, Zhang W, Hu H, Mao X, Zhou L, Yan J, Zhang X, Xu J. Safety and Clinical Response to Combined Immunotherapy with Autologous iNKT Cells and PD-1 +CD8 + T Cells in Patients Failing First-line Chemotherapy in Stage IV Pancreatic Cancer. CANCER RESEARCH COMMUNICATIONS 2023; 3:991-1003. [PMID: 37377605 PMCID: PMC10246506 DOI: 10.1158/2767-9764.crc-23-0137] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/11/2023] [Accepted: 05/15/2023] [Indexed: 06/29/2023]
Abstract
Purpose A phase I clinical trial was conducted to assess the safety and feasibility of invariant natural killer T (iNKT) cells combined with PD-1+CD8+ T cells in patients with advanced pancreatic cancer and failing the first-line chemotherapy. Patients and Methods Fifteen eligible patients were enrolled, of whom 9 received at least three cycles of treatment each. In total, 59 courses were administered. Results Fever was the most common adverse event, peaking at about 2-4 hours after cell infusion and reverting within 24 hours without treatment in all patients. Influenza-like reactions such as headache, myalgia, and arthralgia were also observed in 4, 4, and 3 of the patients, respectively. In addition, vomiting and dizziness were prevalent, while abdominal pain, chest pain, rash, and stuffy nose were rare adverse events, each reported in 1 patient. Side effects above grade 2 were not observed. Two patients achieved partial regression, while 1 patient experienced disease progression assessed 4 weeks after the third course. Three patients are still alive at the time of writing and have progression-free survival longer than 12 months. The overall survival time has been extended to over 12 months in 6 of the 9 patients. No constant changes of CD4+ T, B, and NK cells were recorded except for elevated CD8+ T cells after the first course. Conclusions The combination of autologous iNKT cells and PD-1+CD8+ T cells was a safe therapeutic strategy against advanced pancreatic cancer. The patients exhibited a potentially promising prolonged survival time. Further study appears warranted to evaluate the efficacy of these combined cell infusions in pancreatic cancer. Trial registration This trial was included in the clinical trial which was registered in ClinicalTrials.gov (ID:NCT03093688) on March 15, 2017. Significance There is an unmet need for novel, more effective, and tolerable therapies for pancreatic cancer. Here we present a phase I clinical trial employing iNKT cells combined with PD-1+CD8+ T cells in 9 patients with advanced pancreatic cancer and failing the first-line chemotherapy. The combined immunotherapy was shown to be feasible in the enrolled patients with limited side effects and optimistic clinical responses, which could bring opportunity of therapeutic advancement.
Collapse
Affiliation(s)
- Jing Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Xiaobo Cheng
- Clinical Research Center, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, P.R. China
| | - Yanling Jin
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Bili Xia
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Ran Qin
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Wei Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Huiliang Hu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Xiaoting Mao
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Liting Zhou
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Jia Yan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, P.R. China
- Clinical Center for Biotherapy, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, P.R. China
- Clinical Center for Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Jianqing Xu
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, P.R. China
- Clinical Center for Biotherapy, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, P.R. China
- Clinical Center for Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
9
|
Hovhannisyan L, Riether C, Aebersold DM, Medová M, Zimmer Y. CAR T cell-based immunotherapy and radiation therapy: potential, promises and risks. Mol Cancer 2023; 22:82. [PMID: 37173782 PMCID: PMC10176707 DOI: 10.1186/s12943-023-01775-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
CAR T cell-based therapies have revolutionized the treatment of hematological malignancies such as leukemia and lymphoma within the last years. In contrast to the success in hematological cancers, the treatment of solid tumors with CAR T cells is still a major challenge in the field and attempts to overcome these hurdles have not been successful yet. Radiation therapy is used for management of various malignancies for decades and its therapeutic role ranges from local therapy to a priming agent in cancer immunotherapy. Combinations of radiation with immune checkpoint inhibitors have already proven successful in clinical trials. Therefore, a combination of radiation therapy may have the potential to overcome the current limitations of CAR T cell therapy in solid tumor entities. So far, only limited research was conducted in the area of CAR T cells and radiation. In this review we will discuss the potential and risks of such a combination in the treatment of cancer patients.
Collapse
Affiliation(s)
- Lusine Hovhannisyan
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, Bern, 3008, Switzerland
- Department for Biomedical Research, Radiation Oncology, University of Bern, Murtenstrasse 35, Bern, 3008, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, 3010, Switzerland
| | - Carsten Riether
- Department of Medical Oncology, Inselspital, University Hospital and University of Bern, Bern, 3010, Switzerland
| | - Daniel M Aebersold
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, Bern, 3008, Switzerland
- Department for Biomedical Research, Radiation Oncology, University of Bern, Murtenstrasse 35, Bern, 3008, Switzerland
| | - Michaela Medová
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, Bern, 3008, Switzerland
- Department for Biomedical Research, Radiation Oncology, University of Bern, Murtenstrasse 35, Bern, 3008, Switzerland
| | - Yitzhak Zimmer
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Freiburgstrasse 8, Bern, 3008, Switzerland.
- Department for Biomedical Research, Radiation Oncology, University of Bern, Murtenstrasse 35, Bern, 3008, Switzerland.
| |
Collapse
|
10
|
Yu L, Lanqing G, Huang Z, Xin X, Minglin L, Fa-hui L, Zou H, Min J. T cell immunotherapy for cervical cancer: challenges and opportunities. Front Immunol 2023; 14:1105265. [PMID: 37180106 PMCID: PMC10169584 DOI: 10.3389/fimmu.2023.1105265] [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: 11/22/2022] [Accepted: 02/27/2023] [Indexed: 05/15/2023] Open
Abstract
Cancer cellular immunotherapy has made inspiring therapeutic effects in clinical practices, which brings new hope for the cure of cervical cancer. CD8+T cells are the effective cytotoxic effector cells against cancer in antitumor immunity, and T cells-based immunotherapy plays a crucial role in cellular immunotherapy. Tumor infiltrated Lymphocytes (TIL), the natural T cells, is approved for cervical cancer immunotherapy, and Engineered T cells therapy also has impressive progress. T cells with natural or engineered tumor antigen binding sites (CAR-T, TCR-T) are expanded in vitro, and re-infused back into the patients to eradicate tumor cells. This review summarizes the preclinical research and clinical applications of T cell-based immunotherapy for cervical cancer, and the challenges for cervical cancer immunotherapy.
Collapse
Affiliation(s)
- Lingfeng Yu
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Gong Lanqing
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyu Huang
- School of Arts and Sciences, Brandeis University, Boston, MA, United States
| | - Xiaoyan Xin
- School of Arts and Sciences, Brandeis University, Boston, MA, United States
| | - Liang Minglin
- School of Arts and Sciences, Brandeis University, Boston, MA, United States
| | - Lv Fa-hui
- Department of Obstetrics and Gynecology, The Second People’s Hospital of Hefei, Hefei, Anhui, China
| | - Hongmei Zou
- Department of Obstetrics, Qianjiang Central Hospital, Qianjiang, Hubei, China
| | - Jie Min
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Fudalej M, Kwaśniewska D, Nurzyński P, Badowska-Kozakiewicz A, Mękal D, Czerw A, Sygit K, Deptała A. New Treatment Options in Metastatic Pancreatic Cancer. Cancers (Basel) 2023; 15:cancers15082327. [PMID: 37190255 DOI: 10.3390/cancers15082327] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/12/2023] [Accepted: 04/15/2023] [Indexed: 05/17/2023] Open
Abstract
Pancreatic cancer (PC) is the seventh leading cause of cancer death across the world. Poor prognosis of PC is associated with several factors, such as diagnosis at an advanced stage, early distant metastases, and remarkable resistance to most conventional treatment options. The pathogenesis of PC seems to be significantly more complicated than originally assumed, and findings in other solid tumours cannot be extrapolated to this malignancy. To develop effective treatment schemes prolonging patient survival, a multidirectional approach encompassing different aspects of the cancer is needed. Particular directions have been established; however, further studies bringing them all together and connecting the strengths of each therapy are needed. This review summarises the current literature and provides an overview of new or emerging therapeutic strategies for the more effective management of metastatic PC.
Collapse
Affiliation(s)
- Marta Fudalej
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
- Department of Oncology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | - Daria Kwaśniewska
- Department of Oncology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | - Paweł Nurzyński
- Department of Oncology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | | | - Dominika Mękal
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Aleksandra Czerw
- Department of Health Economics and Medical Law, Medical University of Warsaw, 01-445 Warsaw, Poland
- Department of Economic and System Analyses, National Institute of Public Health NIH-National Research Institute, 00-791 Warsaw, Poland
| | - Katarzyna Sygit
- Faculty of Health Sciences, Calisia University, 62-800 Kalisz, Poland
| | - Andrzej Deptała
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
- Department of Oncology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| |
Collapse
|
12
|
Zhai X, Mao L, Wu M, Liu J, Yu S. Challenges of Anti-Mesothelin CAR-T-Cell Therapy. Cancers (Basel) 2023; 15:cancers15051357. [PMID: 36900151 PMCID: PMC10000068 DOI: 10.3390/cancers15051357] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/10/2023] [Accepted: 02/06/2023] [Indexed: 02/23/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T-cell therapy is a kind of adoptive T-cell therapy (ACT) that has developed rapidly in recent years. Mesothelin (MSLN) is a tumor-associated antigen (TAA) that is highly expressed in various solid tumors and is an important target antigen for the development of new immunotherapies for solid tumors. This article reviews the clinical research status, obstacles, advancements and challenges of anti-MSLN CAR-T-cell therapy. Clinical trials on anti-MSLN CAR-T cells show that they have a high safety profile but limited efficacy. At present, local administration and introduction of new modifications are being used to enhance proliferation and persistence and to improve the efficacy and safety of anti-MSLN CAR-T cells. A number of clinical and basic studies have shown that the curative effect of combining this therapy with standard therapy is significantly better than that of monotherapy.
Collapse
Affiliation(s)
- Xuejia Zhai
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China
| | - Ling Mao
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China
| | - Min Wu
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China
| | - Jie Liu
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China
| | - Shicang Yu
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China
- Jinfeng Laboratory, Chongqing 401329, China
- Correspondence:
| |
Collapse
|
13
|
Molnar A, Monroe H, Basri Aydin H, Arslan ME, Lightle A, Lee H, El Jabbour T. Tumors of the Digestive System: Comprehensive Review of Ancillary Testing and Biomarkers in the Era of Precision Medicine. Curr Oncol 2023; 30:2388-2404. [PMID: 36826143 PMCID: PMC9954843 DOI: 10.3390/curroncol30020182] [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: 01/26/2023] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023] Open
Abstract
Immunotherapy has remained at the vanguard of promising cancer therapeutic regimens due to its exceptionally high specificity for tumor cells and potential for significantly improved treatment-associated quality of life compared to other therapeutic approaches such as surgery and chemoradiation. This is especially true in the digestive system, where high rates of mutation give rise to a host of targetable tumor-specific antigens. Many patients, however, do not exhibit measurable improvements under immunotherapy due to intrinsic or acquired resistance, making predictive biomarkers necessary to determine which patients will benefit from this line of treatment. Many of these biomarkers are assessed empirically by pathologists according to nuanced scoring criteria and algorithms. This review serves to inform clinicians and pathologists of extant and promising upcoming biomarkers predictive of immunotherapeutic efficacy among digestive system malignancies and the ancillary testing required for interpretation by pathologists according to tumor site of origin.
Collapse
Affiliation(s)
- Attila Molnar
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10025, USA
| | - Hunter Monroe
- Department of Pathology, West Virginia University, Morgantown, WV 26506, USA
| | - Hasan Basri Aydin
- Department of Pathology, Albany Medical Center, Albany, NY 12208, USA
| | - Mustafa Erdem Arslan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrea Lightle
- Department of Pathology, Albany Medical Center, Albany, NY 12208, USA
| | - Hwajeong Lee
- Department of Pathology, Albany Medical Center, Albany, NY 12208, USA
| | - Tony El Jabbour
- Department of Pathology, West Virginia University, Morgantown, WV 26506, USA
- Correspondence:
| |
Collapse
|
14
|
Ma Z, Hua J, Liu J, Zhang B, Wang W, Yu X, Xu J. Mesenchymal Stromal Cell-Based Targeted Therapy Pancreatic Cancer: Progress and Challenges. Int J Mol Sci 2023; 24:ijms24043559. [PMID: 36834969 PMCID: PMC9966548 DOI: 10.3390/ijms24043559] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/18/2023] [Accepted: 02/01/2023] [Indexed: 02/12/2023] Open
Abstract
Pancreatic cancer is an aggressive malignancy with high mortality rates and poor prognoses. Despite rapid progress in the diagnosis and treatment of pancreatic cancer, the efficacy of current therapeutic strategies remains limited. Hence, better alternative therapeutic options for treating pancreatic cancer need to be urgently explored. Mesenchymal stromal cells (MSCs) have recently received much attention as a potential therapy for pancreatic cancer owing to their tumor-homing properties. However, the specific antitumor effect of MSCs is still controversial. To this end, we aimed to focus on the potential anti-cancer treatment prospects of the MSC-based approach and summarize current challenges in the clinical application of MSCs to treat pancreatic cancer.
Collapse
Affiliation(s)
- Zhilong Ma
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Jie Hua
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
- Correspondence: (X.Y.); (J.X.); Tel.: +86-021-64175590 (X.Y.); +86-021-64031446 (J.X.)
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
- Correspondence: (X.Y.); (J.X.); Tel.: +86-021-64175590 (X.Y.); +86-021-64031446 (J.X.)
| |
Collapse
|
15
|
Raza F, Evans L, Motallebi M, Zafar H, Pereira-Silva M, Saleem K, Peixoto D, Rahdar A, Sharifi E, Veiga F, Hoskins C, Paiva-Santos AC. Liposome-based diagnostic and therapeutic applications for pancreatic cancer. Acta Biomater 2023; 157:1-23. [PMID: 36521673 DOI: 10.1016/j.actbio.2022.12.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer is one of the harshest and most challenging cancers to treat, often labeled as incurable. Chemotherapy continues to be the most popular treatment yet yields a very poor prognosis. The main barriers such as inefficient drug penetration and drug resistance, have led to the development of drug carrier systems. The benefits, ease of fabrication and modification of liposomes render them as ideal future drug delivery systems. This review delves into the versatility of liposomes to achieve various mechanisms of treatment for pancreatic cancer. Not only are there benefits of loading chemotherapy drugs and targeting agents onto liposomes, as well as mRNA combined therapy, but liposomes have also been exploited for immunotherapy and can be programmed to respond to photothermal therapy. Multifunctional liposomal formulations have demonstrated significant pre-clinical success. Functionalising drug-encapsulated liposomes has resulted in triggered drug release, specific targeting, and remodeling of the tumor environment. Suppressing tumor progression has been achieved, due to their ability to more efficiently and precisely deliver chemotherapy. Currently, no multifunctional surface-modified liposomes are clinically approved for pancreatic cancer thus we aim to shed light on the trials and tribulations and progress so far, with the hope for liposomal therapy in the future and improved patient outcomes. STATEMENT OF SIGNIFICANCE: Considering that conventional treatments for pancreatic cancer are highly associated with sub-optimal performance and systemic toxicity, the development of novel therapeutic strategies holds outmost relevance for pancreatic cancer management. Liposomes are being increasingly considered as promising nanocarriers for providing not only an early diagnosis but also effective, highly specific, and safer treatment, improving overall patient outcome. This manuscript is the first in the last 10 years that revises the advances in the application of liposome-based formulations in bioimaging, chemotherapy, phototherapy, immunotherapy, combination therapies, and emergent therapies for pancreatic cancer management. Prospective insights are provided regarding several advantages resulting from the use of liposome technology in precision strategies, fostering new ideas for next-generation diagnosis and targeted therapies of pancreatic cancer.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Lauren Evans
- Pure and Applied Chemistry, University of Strathclyde, 99 George Street, Glasgow, G1 1RD, UK
| | - Mahzad Motallebi
- Immunology Board for Transplantation And Cell-based Therapeutics (Immuno_TACT), Universal Scientific Education and Research Network (USERN), Tehran 7616911319, Iran; Nanomedicine Research Association (NRA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Miguel Pereira-Silva
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Kalsoom Saleem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 45320, Pakistan
| | - Diana Peixoto
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran
| | - Esmaeel Sharifi
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Clare Hoskins
- Pure and Applied Chemistry, University of Strathclyde, 99 George Street, Glasgow, G1 1RD, UK
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
16
|
Mukherji R, Debnath D, Hartley ML, Noel MS. The Role of Immunotherapy in Pancreatic Cancer. Curr Oncol 2022; 29:6864-6892. [PMID: 36290818 PMCID: PMC9600738 DOI: 10.3390/curroncol29100541] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 01/13/2023] Open
Abstract
Pancreatic adenocarcinoma remains one of the most lethal cancers globally, with a significant need for improved therapeutic options. While the recent breakthroughs of immunotherapy through checkpoint inhibitors have dramatically changed treatment paradigms in other malignancies based on considerable survival benefits, this is not so for pancreatic cancer. Chemotherapies with modest benefits are still the cornerstone of advanced pancreatic cancer treatment. Pancreatic cancers are inherently immune-cold tumors and have been largely refractory to immunotherapies in clinical trials. Understanding and overcoming the current failures of immunotherapy through elucidating resistance mechanisms and developing novel therapeutic approaches are essential to harnessing the potential durable benefits of immune-modulating therapy in pancreatic cancer patients.
Collapse
Affiliation(s)
- Reetu Mukherji
- The Ruesch Center for the Cure of Gastrointestinal Cancers, Georgetown Lombardi Comprehensive Cancer Center, Division of Hematology and Oncology, Medstar Georgetown University Hospital, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Dipanjan Debnath
- Department of Internal Medicine, Medstar Washington Hospital Center, 110 Irving Street NW, Washington, DC 20010, USA
| | - Marion L. Hartley
- The Ruesch Center for the Cure of Gastrointestinal Cancers, Georgetown Lombardi Comprehensive Cancer Center, Division of Hematology and Oncology, Medstar Georgetown University Hospital, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Marcus S. Noel
- The Ruesch Center for the Cure of Gastrointestinal Cancers, Georgetown Lombardi Comprehensive Cancer Center, Division of Hematology and Oncology, Medstar Georgetown University Hospital, 3800 Reservoir Road NW, Washington, DC 20007, USA
- Correspondence:
| |
Collapse
|
17
|
Xue Z, Mei D, Zhang L. Advances in single-cell nanoencapsulation and applications in diseases. J Microencapsul 2022; 39:481-494. [PMID: 35998209 DOI: 10.1080/02652048.2022.2111472] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Single-cell nanoencapsulation is a method of coating the surface of single cell with nanomaterials. In the early 20th century, with the introduction of various types of organic or inorganic nano-polymer materials, the selection of cell types, and the functional modification of the outer coating, this technology has gradually matured. Typical preparation methods include interfacial polycondensation, complex condensation, spray drying, microdroplet ejection, and layer-by-layer (LbL) self-assembly. The LbL assembly technology utilises nanomaterials with opposite charges deposited on cells by strong interaction (electrostatic interaction) or weak interaction (hydrogen bonding, hydrophobic interaction), which drives compounds to spontaneously form films with complete structure, stable performance and unique functions on cells. According to the needs of the disease, choosing appropriate cell types and biocompatible and biodegradable nanomaterials could achieve the purpose of promoting cell proliferation, immune isolation, reducing phagocytosis of the reticuloendothelial system, prolonging the circulation time in vivo, and avoiding repeated administration. Therefore, encapsulated cells could be utilised in various biomedical fields, such as cell catalysis, biotherapy, vaccine manufacturing and antitumor therapy. This article reviews cell nanoencapsulation therapies for diseases, including the various cell sources used, nanoencapsulation technology and the latest advances in preclinical and clinical research.
Collapse
Affiliation(s)
- Ziyang Xue
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei, China
| | - Dan Mei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei, China
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei, China
| |
Collapse
|
18
|
Diverse and precision therapies open new horizons for patients with advanced pancreatic ductal adenocarcinoma. Hepatobiliary Pancreat Dis Int 2022; 21:10-24. [PMID: 34538570 DOI: 10.1016/j.hbpd.2021.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 08/31/2021] [Indexed: 02/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a common cause of cancer-related death, and most patients are with advanced disease when diagnosed. At present, despite a variety of treatments have been developed for PDAC, few effective treatment options are available; on the other hand, PDAC shows significant resistance to chemoradiotherapy, targeted therapy, and immunotherapy due to its heterogeneous genetic profile, molecular signaling pathways, and complex tumor immune microenvironment. Nevertheless, over the past decades, there have been many new advances in the key theory and understanding of the intrinsic mechanisms and complexity of molecular biology and molecular immunology in pancreatic cancer, based on which more and more diverse new means and reasonable combination strategies for PDAC treatment have been developed and preliminary breakthroughs have been made. With the continuous exploration, from surgical local treatment to comprehensive medical management, the research-diagnosis-management system of pancreatic cancer is improving. This review focused on the variety of treatments for advanced PDAC, including traditional chemotherapy, targeted therapy, immunotherapy, microenvironment matrix regulation as well as the treatment targeting epigenetics, metabolism and cancer stem cells. We pointed out the current research bottlenecks and future exploration directions.
Collapse
|
19
|
Wu W, Yang H, Wang Z, Zhang Z, Lu X, Yang W, Xu X, Jiang Y, Li Y, Fan X, Shao Q. A Noncanonical Hedgehog Signaling Exerts a Tumor-Promoting Effect on Pancreatic Cancer Cells Via Induction of Osteopontin Expression. Cancer Biother Radiopharm 2021. [PMID: 34978897 DOI: 10.1089/cbr.2021.0317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Objective: Sonic Hedgehog (Shh)-Gli1 signaling and osteopontin (OPN) play vital roles in pancreatic cancer. However, the precise mechanisms of both signals have not been fully clarified, and whether there is a correlation between them in pancreatic ductal adenocarcinoma (PDAC) is unknown. This study aims to confirm the effect of OPN on human PDAC and assess whether Hh signaling affects pancreatic cancer cells through upregulation of OPN. Materials and Methods: OPN expression in human PDAC tissues and cell lines was investigated. Proliferation, apoptosis, migration, and invasion of OPN-knockdown BxPC-3 cells were observed. We analyzed the correlation between Shh or Gli1 and OPN expression in human PDAC. Hh signaling inhibitors and shRNA against Gli1 were used to confirm if OPN expression in BxPC-3 cells was regulated by Hh canonical or noncanonical pathway. We also evaluated the proliferation, apoptosis, migration, and invasion of Gli1-knockdown BxPC-3 cells. Results: OPN is highly expressed in human PDAC tissues and cell lines. The proliferation, migration, and invasion of BxPC-3 cell lines were decreased, whereas apoptosis was increased when OPN was knocked down. Correlation analysis showed that Gli1, but not Shh, was associated with OPN expression in human PDAC, and Gli1 regulated OPN production in BxPC-3 cells through a noncanonical pathway because Gli but not Smo inhibitor reduced OPN expression. Similar to above, the proliferation, migration, and invasion of BxPC-3 cells were decreased, whereas the apoptosis was increased when Gli1 was knocked down. Supplement of exogenous OPN protein could partially reverse the effect of both OPN knockdown and Gli1 knockdown on the bio-behavior of BxPC-3 cells. Conclusion: Hh signaling promotes proliferation, migration, and invasion but inhibits apoptosis of pancreatic cancer cells through upregulation of OPN in a noncanonical pathway.
Collapse
Affiliation(s)
- Weijiang Wu
- Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, People's Republic of China
- Department of Histology and Embryology, School of Medicine, Jiangsu University, Zhenjiang, People's Republic of China
| | - Hanqing Yang
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, People's Republic of China
- Department of Burns and Plastic Surgery, Wujin Hospital Affiliated to Jiangsu University, Changzhou, People's Republic of China
| | - Zhutao Wang
- Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, People's Republic of China
| | - Zhijian Zhang
- Department of Histology and Embryology, School of Medicine, Jiangsu University, Zhenjiang, People's Republic of China
| | - Xiaodong Lu
- Department of Histology and Embryology, School of Medicine, Jiangsu University, Zhenjiang, People's Republic of China
| | - Wenjing Yang
- Department of Histology and Embryology, School of Medicine, Jiangsu University, Zhenjiang, People's Republic of China
| | - Xiayue Xu
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Yinuo Jiang
- Department of Histology and Embryology, School of Medicine, Jiangsu University, Zhenjiang, People's Republic of China
| | - Yan Li
- Department of Histology and Embryology, School of Medicine, Jiangsu University, Zhenjiang, People's Republic of China
| | - Xin Fan
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Qixiang Shao
- Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, People's Republic of China
- Jiangsu College of Nursing, School of Medical Science and Laboratory Medicine, Huai'an, People's Republic of China
| |
Collapse
|
20
|
Jia Y, Dietrich CF, Sun S. EUS-guided cell transplantation: Planting seeds of hope. Endosc Ultrasound 2021; 10:401-403. [PMID: 34975038 PMCID: PMC8785668 DOI: 10.4103/eus-d-21-00220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Yunbo Jia
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Christoph F Dietrich
- Department of Allgemeine Innere Medizin, Kliniken Hirslanden Bern, Beau Site, Salem and Permanence, Bern, Switzerland
| | - Siyu Sun
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
21
|
Truong LH, Pauklin S. Pancreatic Cancer Microenvironment and Cellular Composition: Current Understandings and Therapeutic Approaches. Cancers (Basel) 2021; 13:5028. [PMID: 34638513 PMCID: PMC8507722 DOI: 10.3390/cancers13195028] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/01/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal human solid tumors, despite great efforts in improving therapeutics over the past few decades. In PDAC, the distinct characteristic of the tumor microenvironment (TME) is the main barrier for developing effective treatments. PDAC TME is characterized by a dense stroma, cancer-associated fibroblasts, and immune cells populations that crosstalk to the subpopulations of neoplastic cells that include cancer stem cells (CSCs). The heterogeneity in TME is also exhibited in the diversity and dynamics of acellular components, including the Extracellular matrix (ECM), cytokines, growth factors, and secreted ligands to signaling pathways. These contribute to drug resistance, metastasis, and relapse in PDAC. However, clinical trials targeting TME components have often reported unexpected results and still have not benefited patients. The failures in those trials and various efforts to understand the PDAC biology demonstrate the highly heterogeneous and multi-faceted TME compositions and the complexity of their interplay within TME. Hence, further functional and mechanistic insight is needed. In this review, we will present a current understanding of PDAC biology with a focus on the heterogeneity in TME and crosstalk among its components. We also discuss clinical challenges and the arising therapeutic opportunities in PDAC research.
Collapse
Affiliation(s)
| | - Siim Pauklin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Old Road, University of Oxford, Oxford OX3 7LD, UK;
| |
Collapse
|
22
|
Bioinformatic Analysis of Prognostic and Immune-Related Genes in Pancreatic Cancer. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:5549298. [PMID: 34394706 PMCID: PMC8355962 DOI: 10.1155/2021/5549298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 06/17/2021] [Accepted: 07/16/2021] [Indexed: 01/21/2023]
Abstract
Pancreatic cancer (PC) is a malignant tumor with poor prognosis. The poor effect of surgery and chemotherapy makes the research of immunotherapy target molecules significant. Therefore, identifying the new molecular targets of PC is important for patients. In our study, we systematically analyzed molecular correlates of pancreatic cancer by bioinformatic analysis. We characterized differentially expressed analysis based on the TCGA pancreatic cancer dataset. Then, univariate Cox regression was employed to screen out overall survival- (OS-) related DEGs. Based on these genes, we established a risk signature by the multivariate Cox regression model. The ICGC cohort and GSE62452 cohort were used to validate the reliability of the risk signature. The impact of T lymphocyte-related genes from risk signature was confirmed in PC. Here, we observed the correlation between the T lymphocyte-related genes and the expression level of targeted therapy. We established a five-mRNA (LY6D, ANLN, ZNF488, MYEOV, and SCN11A) prognostic risk signature. Next, we identified ANLN and MYEOV that were associated with T lymphocyte infiltrations (P < 0.05). High ANLN and MYEOV expression levels had a poorer prognosis in decreased T lymphocyte subgroup in PC. Correlation analysis between ANLN and MYEOV and immunomodulators showed that ANLN and MYEOV may have potential value in pancreatic cancer immunotherapy.
Collapse
|
23
|
Galanopoulos M, Doukatas A, Gkeros F, Viazis N, Liatsos C. Room for improvement in the treatment of pancreatic cancer: Novel opportunities from gene targeted therapy. World J Gastroenterol 2021; 27:3568-3580. [PMID: 34239270 PMCID: PMC8240062 DOI: 10.3748/wjg.v27.i24.3568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/11/2021] [Accepted: 05/22/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the highest and in fact, unchanged mortality-associated tumor, with an exceptionally low survival rate due to its challenging diagnostic approach. So far, its treatment is based on a combination of approaches (such as surgical resection with or rarely without chemotherapeutic agents), but with finite limits. Thus, looking for additional space to improve pancreatic tumorigenesis therapeutic approach, research has focused on gene therapy with unexpectedly growing horizons not only for the treatment of inoperable pancreatic disease, but also for its early stages. In vivo gene delivery viral vectors, despite few disadvantages (possible immunogenicity, toxicity, mutagenicity, or high cost), could be one of the most efficient cancer gene therapeutic strategies for clinical application due to their superiority compared with other systems (ex vivo delivery strategies). Their dominance consists of simple preparation, easy operation and a wide range of functions. Adenoviruses are one of the most common used vectors, inducing strong immune as well as inflammatory reactions. Oncolytic virotherapy, using the above mentioned in vivo viral vectors, is one of the most promising non-pathogenic, highly-selective cytotoxic anti-cancer therapy using anti-cancer agents with high anti-tumor potency and strong oncolytic effect. There have been a variety of targeted therapeutic and pre-clinical strategies tested for gene therapy in pancreatic cancer such as gene-editing systems (e.g., clustered regularly interspaced palindromic repeats-Cas9), RNA interference technology (e.g., microRNAs, short hairpin RNA or small interfering RNA), adoptive immunotherapy and vaccination (e.g., chimeric antigen receptor T-cell therapy) with encouraging results.
Collapse
Affiliation(s)
- Michail Galanopoulos
- Department of Gastroenterology, Addenbrooke’s Hospital, Cambridge CB2 0QQ, United Kingdom
| | - Aris Doukatas
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens GR 15772, Greece
| | - Filippos Gkeros
- Department of Gastroenterology, Evangelismos, Ophthalmiatreion Athinon and Polyclinic Hospitals, Athens 10676, Greece
| | - Nikos Viazis
- Department of Gastroenterology, Evangelismos, Ophthalmiatreion Athinon and Polyclinic Hospitals, Athens 10676, Greece
| | - Christos Liatsos
- Department of Gastroenterology, 401 General Military Hospital, Athens 11525, Greece
| |
Collapse
|
24
|
Anwar MY, Williams GR, Paluri RK. CAR T Cell Therapy in Pancreaticobiliary Cancers: a Focused Review of Clinical Data. J Gastrointest Cancer 2021; 52:1-10. [PMID: 32700185 DOI: 10.1007/s12029-020-00457-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE CAR T cell therapy is an innovative approach to treat cancers in the modern era. It utilizes the application of chimeric antigen receptors targeted against specific antigens expressed by the tumor cells. Although its efficacy is established in hematological malignancies, the safety and efficacy of this therapy in solid tumors, especially pancreaticobiliary cancers, is a highly investigated aspect. A focused review of clinical data was conducted to examine the outcomes of this therapy in pancreaticobiliary cancers. METHODS A comprehensive literature search was done on Medline and Embase databases through April 24, 2020 for studies that evaluated the outcomes of CAR T cell therapy in pancreaticobiliary cancers. RESULTS There were six phase 1 trials, while one was phase 1/2. Some of these trials were specifically done for pancreaticobiliary cancers, while others included patients of various solid organ cancers, including pancreatic and biliary tract cancers. The target antigens for therapy in these trials included mesothelin, CD133, prostate stem cell antigen, claudin 18.2, epidermal growth factor receptor, and human epidermal growth factor receptor 2. CAR T cell therapy has shown very few grade 3 and 4 side effects. Most of the adverse events are associated with cytokine release syndrome. CONCLUSION CAR T cell therapy has a manageable safety profile based on phase 1 studies, and efficacy assessments are currently ongoing in dose expansion and phase 2 studies.
Collapse
Affiliation(s)
| | - Grant R Williams
- O'Neil Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ravi K Paluri
- O'Neil Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
25
|
Preparation of CD3 Antibody-Conjugated, Graphene Oxide Coated Iron Nitride Magnetic Beads and Its Preliminary Application in T Cell Separation. MAGNETOCHEMISTRY 2021. [DOI: 10.3390/magnetochemistry7050058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immunomagnetic beads (IMBs) for cell sorting are universally used in medical and biological fields. At present, the IMBs on the market are ferrite coated with a silicon shell. Based on a new type of magnetic material, the graphene coated iron nitride magnetic particle (G@FeN-MP), which we previously reported, we prepared a novel IMB, a graphene oxide coated iron nitride immune magnetic bead (GO@FeN-IMBs), and explored its feasibility for cell sorting. First, the surface of the G@FeN-MP was oxidized to produce oxygen-containing groups as carboxyl, etc. by the optimized Hummers’ method, followed by a homogenization procedure to make the particles uniform in size and dispersive. The carboxy groups generated were then condensed and coupled with anti-CD3 antibodies by the carbodiimide method to produce an anti-CD3-GO@FeN-IMB after the coupling efficacy was proved by bovine serum albumin (BSA) and labeled antibodies. Finally, the anti-CD3-GO@FeN-IMBs were incubated with a cell mixture containing human T cells. With the aid of a magnetic stand, the T cells were successfully isolated from the cell mixture. The isolated T cells turned out to be intact and could proliferate with the activation of the IMBs. The results show that the G@FeN-MP can be modified for IMB preparation, and the anti-CD3-GO@FeN-IMBs we prepared can potentially separate T cells.
Collapse
|
26
|
Elsayed M, Abdelrahim M. The Latest Advancement in Pancreatic Ductal Adenocarcinoma Therapy: A Review Article for the Latest Guidelines and Novel Therapies. Biomedicines 2021; 9:389. [PMID: 33917380 PMCID: PMC8067364 DOI: 10.3390/biomedicines9040389] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer deaths in the US, and it is expected to be the second leading cause of cancer deaths by 2030. The lack of effective early screening tests and alarming symptoms with early undetectable micro-metastasis at the time of presentation play a vital role in the high death rate from pancreatic cancer. In addition to this, the low mutation burden in pancreatic cancer, low immunological profile, dense tumorigenesis stroma, and decreased tumor sensitivity to cytotoxic drugs contribute to the low survival rates in PDAC patients. Despite breakthroughs in chemotherapeutic and immunotherapeutic drugs, pancreatic cancer remains one of the solid tumors that exhibit meager curative rates. Therefore, researchers must dedicate more effort to understanding the pathology and immunological behavior of PDAC, in addition to properly utilizing more advanced screening modalities and new therapeutic agents. In our review, we focus mainly on the latest updates from clinical guidelines and novel therapies that have been recently investigated or are under investigation for PDAC. We used PubMed as a search tool for finding original research articles addressing the latest developments in diagnosing and treating PDAC. Additionally, we also used the clinical trials published on clinicaltrialsgov as sources for our data.
Collapse
Affiliation(s)
- Marwa Elsayed
- School of Medicine, University of Missouri Kansas City, 2301 Holmes, St. Kansas City, MO 64018, USA;
| | - Maen Abdelrahim
- Houston Methodist Cancer Center, Houston Methodist Hospital, 6445 Main Street, Outpatient Center, 24th Floor, Houston, TX 77030, USA
- Cockrell Center of Advanced Therapeutics Phase I Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Weill Cornell Medical College, Institute of Academic Medicine, Houston, TX 77030, USA
| |
Collapse
|
27
|
Wang W, Xu Z, Wang N, Yao R, Qin T, Lin H, Yue L. Prognostic value of eight immune gene signatures in pancreatic cancer patients. BMC Med Genomics 2021; 14:42. [PMID: 33546693 PMCID: PMC7863419 DOI: 10.1186/s12920-020-00868-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 12/29/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pancreatic cancer is one of the most common malignant tumors of the digestive tract, and it has a poor prognosis. Traditional methods are not effective to accurately assess the prognosis of patients with pancreatic cancer. Immunotherapy is a new promising approach for the treatment of pancreatic cancer; however, some patients do not respond well to immunotherapy, which may be related to tumor microenvironment regulation. In this study, we use gene expression database to mine important immune genes and establish a prognostic prediction model for pancreatic cancer patients. We hope to provide a feasible method to evaluate the prognosis of pancreatic cancer and provide valuable targets for pancreatic cancer immunotherapy. RESULTS We used univariate COX proportional hazard regression analysis, the least absolute shrinkage and selection operator, and multivariate COX regression analysis to screen 8 genes related to prognosis from the 314 immune-related genes, and used them to construct a new clinical prediction model in the TCGA pancreatic cancer cohort. Subsequently, we evaluated the prognostic value of the model. The Kaplan-Meier cumulative curve showed that patients with low risk scores survived significantly longer than patients with high risk scores. The area under the ROC curve (AUC value) of the risk score was 0.755. The univariate COX analysis showed that the risk score was significantly related to overall survival (HR 1.406, 95% CI 1.237-1.598, P < 0.001), and multivariate analysis showed that the risk score was an independent prognostic factor (HR 1.400, 95% CI 1.287-1.522, P < 0.001). Correlation analysis found that immune genes are closely related to tumor immune microenvironment. CONCLUSIONS Based on the TCGA-PAAD cohort, we identified immune-related markers with independent prognostic significance, validated, and analyzed their biological functions, to provide a feasible method for the prognosis of pancreatic cancer and provide potentially valuable targets for pancreatic cancer immunotherapy.
Collapse
Affiliation(s)
- Wenting Wang
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, 5 Donghaizhong Road, Qingdao, 266071, Shandong, People's Republic of China
| | - Zhijian Xu
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, 5 Donghaizhong Road, Qingdao, 266071, Shandong, People's Republic of China
| | - Ning Wang
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, 5 Donghaizhong Road, Qingdao, 266071, Shandong, People's Republic of China
| | - Ruyong Yao
- Department of Central Laboratory, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong, People's Republic of China
| | - Tao Qin
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, 5 Donghaizhong Road, Qingdao, 266071, Shandong, People's Republic of China
| | - Hao Lin
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, 5 Donghaizhong Road, Qingdao, 266071, Shandong, People's Republic of China
| | - Lu Yue
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, 5 Donghaizhong Road, Qingdao, 266071, Shandong, People's Republic of China.
| |
Collapse
|
28
|
Garcia-Sampedro A, Gaggia G, Ney A, Mahamed I, Acedo P. The State-of-the-Art of Phase II/III Clinical Trials for Targeted Pancreatic Cancer Therapies. J Clin Med 2021; 10:566. [PMID: 33546207 PMCID: PMC7913382 DOI: 10.3390/jcm10040566] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is a devastating disease with very poor prognosis. Currently, surgery followed by adjuvant chemotherapy represents the only curative option which, unfortunately, is only available for a small group of patients. The majority of pancreatic cancer cases are diagnosed at advanced or metastatic stage when surgical resection is not possible and treatment options are limited. Thus, novel and more effective therapeutic strategies are urgently needed. Molecular profiling together with targeted therapies against key hallmarks of pancreatic cancer appear as a promising approach that could overcome the limitations of conventional chemo- and radio-therapy. In this review, we focus on the latest personalised and multimodal targeted therapies currently undergoing phase II or III clinical trials. We discuss the most promising findings of agents targeting surface receptors, angiogenesis, DNA damage and cell cycle arrest, key signalling pathways, immunotherapies, and the tumour microenvironment.
Collapse
Affiliation(s)
| | | | | | | | - Pilar Acedo
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, London NW3 2QG, UK; (A.G.-S.); (G.G.); (A.N.); (I.M.)
| |
Collapse
|
29
|
Abstract
Pancreatic cancer is a tumor with a high degree of malignancy, morbidity, and mortality. Immunotherapy is another important treatment for pancreatic cancer in addition to surgery and chemotherapy, but its application in pancreatic cancer is very limited, which is related to the unique biological behavior of pancreatic cancer and the tumor microenvironment. The immunosuppressive microenvironment of pancreatic cancer is highly heterogeneous and presents challenges for immunotherapy. The transformation of tumor immunosuppressive microenvironment contributes to the response to tumor immunotherapy, such that the tumor undergoes functional reprogramming to change from immunologically "cold" to immunologically "hot." In this review, we summarized the research and progress in immunotherapy for pancreatic cancer, including immune checkpoint inhibitors, vaccines, adoptive T cell therapy, oncolytic viruses, and immunomodulators, and suggest that individualized, combination, and precise therapy should be the main direction of future immunotherapy in pancreatic cancer.
Collapse
Affiliation(s)
- Jia Wu
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang Province, China
| | - Jianting Cai
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang Province, China.
| |
Collapse
|
30
|
Yang JS, Wang CC, Qiu JD, Ren B, You L. Arginine metabolism: a potential target in pancreatic cancer therapy. Chin Med J (Engl) 2020; 134:28-37. [PMID: 33395072 PMCID: PMC7862822 DOI: 10.1097/cm9.0000000000001216] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
ABSTRACT Pancreatic ductal adenocarcinoma (PDAC) is an extremely malignant disease, which has an extremely low survival rate of <9% in the United States. As a new hallmark of cancer, metabolism reprogramming exerts crucial impacts on PDAC development and progression. Notably, arginine metabolism is altered in PDAC cells and participates in vital signaling pathways. In addition, arginine and its metabolites including polyamine, creatine, agmatine, and nitric oxide regulate the proliferation, growth, autophagy, apoptosis, and metastasis of cancer cells. Due to the loss of argininosuccinate synthetase 1 (ASS1) expression, the key enzyme in arginine biosynthesis, arginine deprivation is regarded as a potential strategy for PDAC therapy. However, drug resistance develops during arginine depletion treatment, along with the re-expression of ASS1, metabolic dysfunction, and the appearance of anti-drug antibody. Additionally, arginase 1 exerts crucial roles in myeloid-derived suppressor cells, indicating its potential targeting by cancer immunotherapy. In this review, we introduce arginine metabolism and its impacts on PDAC cells. Also, we discuss the role of arginine metabolism in arginine deprivation therapy and immunotherapy for cancer.
Collapse
Affiliation(s)
- Jin-Shou Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | | | | | | | | |
Collapse
|
31
|
Novel cellular immunotherapy using NKG2D CAR-T for the treatment of cervical cancer. Biomed Pharmacother 2020; 131:110562. [PMID: 32920508 DOI: 10.1016/j.biopha.2020.110562] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/16/2020] [Accepted: 07/26/2020] [Indexed: 12/21/2022] Open
Abstract
Cellular immunotherapy, including chimeric antigen receptor (CAR) modified T cell therapy, has been regarded as one of the most potential antineoplastic drugs for hematological malignancies and solid tumor. However, due to lacking the suitable target, there is no CAR-T drug had been appoved by FDA for the treatment of cervical cancer, one of the most malignant cancers for women. In current study, we designed a NKG2D CAR-T targeting NKG2DL. The NKG2D CAR-T exhibited high-efficient anti-tumor capacity for NKG2DL positive cervical cancer cell line in vitro. In addition, the amount of cytokines secreted from CAR-T cells have had significantly enhanced after co-cultured with NKG2DL positive tumor cell in vitro. In vivo, NKG2D CAR-T cells presented a robust capacity of significantly suppressing tumor growth. Moreover, there was no obvious off-target toxicity after NKG2D CAR-T infusion. Taken together, NKG2D CAR-T showed excellent therapy effect for cervical cancer and might be used as a novel cellular therapeutic agent for treating cervical cancer.
Collapse
|
32
|
Saka D, Gökalp M, Piyade B, Cevik NC, Arik Sever E, Unutmaz D, Ceyhan GO, Demir IE, Asimgil H. Mechanisms of T-Cell Exhaustion in Pancreatic Cancer. Cancers (Basel) 2020; 12:cancers12082274. [PMID: 32823814 PMCID: PMC7464444 DOI: 10.3390/cancers12082274] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
T-cell exhaustion is a phenomenon that represents the dysfunctional state of T cells in chronic infections and cancer and is closely associated with poor prognosis in many cancers. The endogenous T-cell immunity and genetically edited cell therapies (CAR-T) failed to prevent tumor immune evasion. The effector T-cell activity is perturbed by an imbalance between inhibitory and stimulatory signals causing a reprogramming in metabolism and the high levels of multiple inhibitory receptors like programmed cell death protein-1 (PD-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), and Lymphocyte-activation gene 3 (Lag-3). Despite the efforts to neutralize inhibitory receptors by a single agent or combinatorial immune checkpoint inhibitors to boost effector function, PDAC remains unresponsive to these therapies, suggesting that multiple molecular mechanisms play a role in stimulating the exhaustion state of tumor-infiltrating T cells. Recent studies utilizing transcriptomics, mass cytometry, and epigenomics revealed a critical role of Thymocyte selection-associated high mobility group box protein (TOX) genes and TOX-associated pathways, driving T-cell exhaustion in chronic infection and cancer. Here, we will review recently defined molecular, genetic, and cellular factors that drive T-cell exhaustion in PDAC. We will also discuss the effects of available immune checkpoint inhibitors and the latest clinical trials targeting various molecular factors mediating T-cell exhaustion in PDAC.
Collapse
Affiliation(s)
- Didem Saka
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
| | - Muazzez Gökalp
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
| | - Betül Piyade
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
| | - Nedim Can Cevik
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
| | - Elif Arik Sever
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
| | - Derya Unutmaz
- Jackson Laboratory of Genomic Medicine, Farmington, CT 06032, USA;
| | - Güralp O. Ceyhan
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
- Correspondence: (G.O.C.); (I.E.D.); Tel.: +90-5320514424 (G.O.C.); +49-8941405868 (I.E.D.)
| | - Ihsan Ekin Demir
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
- Department of Surgery, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- Correspondence: (G.O.C.); (I.E.D.); Tel.: +90-5320514424 (G.O.C.); +49-8941405868 (I.E.D.)
| | - Hande Asimgil
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
- Department of Surgery, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
33
|
Xie M, Liu D, Yang Y. Anti-cancer peptides: classification, mechanism of action, reconstruction and modification. Open Biol 2020; 10:200004. [PMID: 32692959 PMCID: PMC7574553 DOI: 10.1098/rsob.200004] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Anti-cancer peptides (ACPs) are a series of short peptides composed of 10-60 amino acids that can inhibit tumour cell proliferation or migration, or suppress the formation of tumour blood vessels, and are less likely to cause drug resistance. The aforementioned merits make ACPs the most promising anti-cancer candidate. However, ACPs may be degraded by proteases, or result in cytotoxicity in many cases. To overcome these drawbacks, a plethora of research has focused on reconstruction or modification of ACPs to improve their anti-cancer activity, while reducing their cytotoxicity. The modification of ACPs mainly includes main chain reconstruction and side chain modification. After summarizing the classification and mechanism of action of ACPs, this paper focuses on recent development and progress about their reconstruction and modification. The information collected here may provide some ideas for further research on ACPs, in particular their modification.
Collapse
Affiliation(s)
- Mingfeng Xie
- Department of Bioengineering, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519040, People's Republic of China
| | - Dijia Liu
- Department of Bioengineering, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519040, People's Republic of China
| | - Yufeng Yang
- Department of Bioengineering, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519040, People's Republic of China.,Zhuhai Key Laboratory of Fundamental and Applied Research in Traditional Chinese Medicine, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519040, People's Republic of China
| |
Collapse
|
34
|
Mundry CS, Eberle KC, Singh PK, Hollingsworth MA, Mehla K. Local and systemic immunosuppression in pancreatic cancer: Targeting the stalwarts in tumor's arsenal. Biochim Biophys Acta Rev Cancer 2020; 1874:188387. [PMID: 32579889 DOI: 10.1016/j.bbcan.2020.188387] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023]
Abstract
Late detection, compromised immune system, and chemotherapy resistance underlie the poor patient prognosis for pancreatic ductal adenocarcinoma (PDAC) patients, making it the 3rd leading cause of cancer-related deaths in the United States. Cooperation between the tumor cells and the immune system leads to the immune escape and eventual establishment of the tumor. For more than 20 years, sincere efforts have been made to intercept the tumor-immune crosstalk and identify the probable therapeutic targets for breaking self-tolerance toward tumor antigens. However, the success of these studies depends on detailed examination and understanding of tumor-immune cell interactions, not only in the primary tumor but also at distant systemic niches. Innate and adaptive arms of the immune system sculpt tumor immunogenicity, where they not only aid in providing an amenable environment for their survival but also act as a driver for tumor relapse at primary or distant organ sites. This review article highlights the key events associated with tumor-immune communication and associated immunosuppression at both local and systemic microenvironments in PDAC. Furthermore, we discuss the approaches and benefits of targeting both local and systemic immunosuppression for PDAC patients. The present articles integrate data from clinical and genetic mouse model studies to provide a widespread consensus on the role of local and systemic immunosuppression in undermining the anti-tumor immune responses against PDAC.
Collapse
MESH Headings
- Adaptive Immunity/drug effects
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Bone Marrow/drug effects
- Bone Marrow/immunology
- Bone Marrow/pathology
- Cancer Vaccines/administration & dosage
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/therapy
- Chemotherapy, Adjuvant/methods
- Clinical Trials as Topic
- Combined Modality Therapy/methods
- Disease Models, Animal
- Disease-Free Survival
- Fluorouracil/pharmacology
- Fluorouracil/therapeutic use
- Humans
- Immunity, Innate/drug effects
- Immunotherapy/methods
- Irinotecan/pharmacology
- Irinotecan/therapeutic use
- Leucovorin/pharmacology
- Leucovorin/therapeutic use
- Lymph Node Excision
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Lymph Nodes/surgery
- Mice
- Mice, Transgenic
- Neoadjuvant Therapy/methods
- Oxaliplatin/pharmacology
- Oxaliplatin/therapeutic use
- Pancreas/immunology
- Pancreas/pathology
- Pancreas/surgery
- Pancreatectomy
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/mortality
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- Spleen/immunology
- Spleen/pathology
- Spleen/surgery
- Splenectomy
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Transplantation, Autologous/methods
- Tumor Escape/drug effects
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
- United States/epidemiology
Collapse
Affiliation(s)
- Clara S Mundry
- The Eppley Institute for Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Kirsten C Eberle
- The Eppley Institute for Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Pankaj K Singh
- The Eppley Institute for Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA; Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA; Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Michael A Hollingsworth
- The Eppley Institute for Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA; Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Kamiya Mehla
- The Eppley Institute for Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA.
| |
Collapse
|
35
|
Mucciolo G, Roux C, Scagliotti A, Brugiapaglia S, Novelli F, Cappello P. The dark side of immunotherapy: pancreatic cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:491-520. [PMID: 35582441 PMCID: PMC8992483 DOI: 10.20517/cdr.2020.13] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/23/2020] [Accepted: 04/07/2020] [Indexed: 12/13/2022]
Abstract
Since the journal Science deemed cancer immunotherapy as the "breakthrough of the year" in 2014, there has been an explosion of clinical trials involving immunotherapeutic approaches that, in the last decade - thanks also to the renaissance of the immunosurveillance theory (renamed the three Es theory) - have been continuously and successfully developed. In the latest update of the development of the immuno-oncology drug pipeline, published last November by Nature Review Drug Discovery, it was clearly reported that the immunoactive drugs under study almost doubled in just two years. Of the different classes of passive and active immunotherapies, "cell therapy" is the fastest growing. The aim of this review is to discuss the preclinical and clinical studies that have focused on different immuno-oncology approaches applied to pancreatic cancer, which we assign to the "dark side" of immunotherapy, in the sense that it represents one of the solid tumors showing less response to this type of therapeutic strategy.
Collapse
Affiliation(s)
- Gianluca Mucciolo
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza di Torino, Turin 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10126, Italy
- The two authors contributed equally
| | - Cecilia Roux
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza di Torino, Turin 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10126, Italy
- The two authors contributed equally
| | - Alessandro Scagliotti
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza di Torino, Turin 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10126, Italy
| | - Silvia Brugiapaglia
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza di Torino, Turin 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10126, Italy
| | - Francesco Novelli
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza di Torino, Turin 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10126, Italy
- Molecular Biotechnology Center, University of Turin, Turin 10126, Italy
| | - Paola Cappello
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza di Torino, Turin 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10126, Italy
- Molecular Biotechnology Center, University of Turin, Turin 10126, Italy
| |
Collapse
|
36
|
Motwani K, Peters LD, Vliegen WH, El-sayed AG, Seay HR, Lopez MC, Baker HV, Posgai AL, Brusko MA, Perry DJ, Bacher R, Larkin J, Haller MJ, Brusko TM. Human Regulatory T Cells From Umbilical Cord Blood Display Increased Repertoire Diversity and Lineage Stability Relative to Adult Peripheral Blood. Front Immunol 2020; 11:611. [PMID: 32351504 PMCID: PMC7174770 DOI: 10.3389/fimmu.2020.00611] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 03/17/2020] [Indexed: 12/22/2022] Open
Abstract
The human T lymphocyte compartment is highly dynamic over the course of a lifetime. Of the many changes, perhaps most notable is the transition from a predominantly naïve T cell state at birth to the acquisition of antigen-experienced memory and effector subsets following environmental exposures. These phenotypic changes, including the induction of T cell exhaustion and senescence, have the potential to negatively impact efficacy of adoptive T cell therapies (ACT). When considering ACT with CD4+CD25+CD127-/lo regulatory T cells (Tregs) for the induction of immune tolerance, we previously reported ex vivo expanded umbilical cord blood (CB) Tregs remained more naïve, suppressed responder T cells equivalently, and exhibited a more diverse T cell receptor (TCR) repertoire compared to expanded adult peripheral blood (APB) Tregs. Herein, we hypothesized that upon further characterization, we would observe increased lineage heterogeneity and phenotypic diversity in APB Tregs that might negatively impact lineage stability, engraftment capacity, and the potential for Tregs to home to sites of tissue inflammation following ACT. We compared the phenotypic profiles of human Tregs isolated from CB versus the more traditional source, APB. We conducted analysis of fresh and ex vivo expanded Treg subsets at both the single cell (scRNA-seq and flow cytometry) and bulk (microarray and cytokine profiling) levels. Single cell transcriptional profiles of pre-expansion APB Tregs highlighted a cluster of cells that showed increased expression of genes associated with effector and pro-inflammatory phenotypes (CCL5, GZMK, CXCR3, LYAR, and NKG7) with low expression of Treg markers (FOXP3 and IKZF2). CB Tregs were more diverse in TCR repertoire and homogenous in phenotype, and contained fewer effector-like cells in contrast with APB Tregs. Interestingly, expression of canonical Treg markers, such as FOXP3, TIGIT, and IKZF2, were increased in CB CD4+CD127+ conventional T cells (Tconv) compared to APB Tconv, post-expansion, implying perinatal T cells may adopt a default regulatory program. Collectively, these data identify surface markers (namely CXCR3) that could be depleted to improve purity and stability of APB Tregs, and support the use of expanded CB Tregs as a potentially optimal ACT modality for the treatment of autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Keshav Motwani
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Leeana D. Peters
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Willem H. Vliegen
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Ahmed Gomaa El-sayed
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Howard R. Seay
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - M. Cecilia Lopez
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Henry V. Baker
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Amanda L. Posgai
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Maigan A. Brusko
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Daniel J. Perry
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Rhonda Bacher
- Department of Biostatistics, University of Florida, Gainesville, FL, United States
| | - Joseph Larkin
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Michael J. Haller
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Todd M. Brusko
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
37
|
Tomás-Bort E, Kieler M, Sharma S, Candido JB, Loessner D. 3D approaches to model the tumor microenvironment of pancreatic cancer. Theranostics 2020; 10:5074-5089. [PMID: 32308769 PMCID: PMC7163433 DOI: 10.7150/thno.42441] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/19/2020] [Indexed: 01/18/2023] Open
Abstract
In tumor engineering, 3D approaches are used to model components of the tumor microenvironment and to test new treatments. Pancreatic cancers are a cancer of substantial unmet need and survival rates are lower compared to any other cancer. Bioengineering techniques are increasingly applied to understand the unique biology of pancreatic tumors and to design patient-specific models. Here we summarize how extracellular and cellular elements of the pancreatic tumor microenvironment and their interactions have been studied in 3D cell cultures. We review selected clinical trials, assess the benefits of therapies interfering with the tumor microenvironment and address their limitations and future perspectives.
Collapse
|
38
|
Fan JQ, Wang MF, Chen HL, Shang D, Das JK, Song J. Current advances and outlooks in immunotherapy for pancreatic ductal adenocarcinoma. Mol Cancer 2020; 19:32. [PMID: 32061257 PMCID: PMC7023714 DOI: 10.1186/s12943-020-01151-3] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 02/06/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an incurable cancer resistant to traditional treatments, although a limited number of early-stage patients can undergo radical resection. Immunotherapies for the treatment of haematological malignancies as well as solid tumours have been substantially improved over the past decades, and impressive results have been obtained in recent preclinical and clinical trials. However, PDAC is likely the exception because of its unique tumour microenvironment (TME). In this review, we summarize the characteristics of the PDAC TME and focus on the network of various tumour-infiltrating immune cells, outlining the current advances in PDAC immunotherapy and addressing the effect of the PDAC TME on immunotherapy. This review further explores the combinations of different therapies used to enhance antitumour efficacy or reverse immunodeficiencies and describes optimizable immunotherapeutic strategies for PDAC. The concordant combination of various treatments, such as targeting cancer cells and the stroma, reversing suppressive immune reactions and enhancing antitumour reactivity, may be the most promising approach for the treatment of PDAC. Traditional treatments, especially chemotherapy, may also be optimized for individual patients to remodel the immunosuppressive microenvironment for enhanced therapy.
Collapse
Affiliation(s)
- Jia-qiao Fan
- Third General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Meng-Fei Wang
- Third General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hai-Long Chen
- Third General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Third General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jugal K. Das
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, TX USA
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, TX USA
| |
Collapse
|
39
|
Nevala-Plagemann C, Hidalgo M, Garrido-Laguna I. From state-of-the-art treatments to novel therapies for advanced-stage pancreatic cancer. Nat Rev Clin Oncol 2020; 17:108-123. [PMID: 31705130 DOI: 10.1038/s41571-019-0281-6] [Citation(s) in RCA: 236] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2019] [Indexed: 12/13/2022]
Abstract
Improvements in the outcomes of patients with pancreatic ductal adenocarcinoma (PDAC) have lagged behind advances made in the treatment of many other malignancies over the past few decades. For most patients with PDAC, cytotoxic chemotherapy remains the mainstay of treatment. For patients with resectable disease, modified 5-fluorouracil, leucovorin, irinotecan and oxaliplatin (mFOLFIRINOX) is the standard-of-care adjuvant therapy, although data from several randomized trials have shown improved outcomes with neoadjuvant treatment strategies. For patients with advanced-stage or metastatic disease, comprehensive genomic profiling has revealed several potentially actionable alterations in small subsets of patients and the feasibility of implementing such strategies is beginning to be confirmed. Novel therapies targeting certain aberrations, most notably BRCA1/2 mutations, mismatch repair (MMR) deficiencies or NTRK1-3 fusions, have shown considerable activity in clinical trials, and larotrectinib, entrectinib and pembrolizumab have received FDA approval for the treatment of patients with tumours harbouring NTRK fusions and MMR deficiencies, respectively, regardless of primary tumour histology. In this Review, we describe the available data on the activity of these and other agents in patients with PDAC. Our discussion is structured according to the acronym 'PRIME' to organize the various treatment strategies currently undergoing evaluation in clinical trials: Pathway inhibition, alteration of DNA Repair pathways, Immunotherapy, cancer Metabolism and targeting the Extracellular tumour microenvironment.
Collapse
Affiliation(s)
| | - Manuel Hidalgo
- Division of Hematology and Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Ignacio Garrido-Laguna
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA.
- Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
40
|
Sur D, Havasi A, Cainap C, Samasca G, Burz C, Balacescu O, Lupan I, Deleanu D, Irimie A. Chimeric Antigen Receptor T-Cell Therapy for Colorectal Cancer. J Clin Med 2020; 9:jcm9010182. [PMID: 31936611 PMCID: PMC7019711 DOI: 10.3390/jcm9010182] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/03/2020] [Accepted: 01/07/2020] [Indexed: 02/06/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents a new genetically engineered method of immunotherapy for cancer. The patient’s T-cells are modified to express a specific receptor that sticks to the tumor antigen. This modified cell is then reintroduced into the patient’s body to fight the resilient cancer cells. After exhibiting positive results in hematological malignancies, this therapy is being proposed for solid tumors like colorectal cancer. The clinical data of CAR T-cell therapy in colorectal cancer is rather scarce. In this review, we summarize the current state of knowledge, challenges, and future perspectives of CAR T-cell therapy in colorectal cancer. A total of 22 articles were included in this review. Eligible studies were selected and reviewed by two researchers from 49 articles found on Pubmed, Web of Science, and clinicaltrials.gov. This therapy, at the moment, provides modest benefits in solid tumors. Not taking into consideration the high manufacturing and retail prices, there are still limitations like increased toxicities, relapses, and unfavorable tumor microenvironment for CAR T-cell therapy in colorectal cancer.
Collapse
Affiliation(s)
- Daniel Sur
- 11th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400015 Cluj-Napoca, Romania; (D.S.); (C.C.); (O.B.)
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania; (A.H.); (C.B.)
| | - Andrei Havasi
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania; (A.H.); (C.B.)
| | - Calin Cainap
- 11th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400015 Cluj-Napoca, Romania; (D.S.); (C.C.); (O.B.)
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania; (A.H.); (C.B.)
| | - Gabriel Samasca
- Department of Immunology and Allergology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400162 Cluj-Napoca, Romania;
- Correspondence:
| | - Claudia Burz
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania; (A.H.); (C.B.)
- Department of Immunology and Allergology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400162 Cluj-Napoca, Romania;
| | - Ovidiu Balacescu
- 11th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400015 Cluj-Napoca, Romania; (D.S.); (C.C.); (O.B.)
- Department of Functional Genomics, Proteomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania
| | - Iulia Lupan
- Department of Molecular Biology and Biotehnology, Babeș-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Diana Deleanu
- Department of Immunology and Allergology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400162 Cluj-Napoca, Romania;
| | - Alexandru Irimie
- 11th Department of Oncological Surgery and Gynecological Oncology, “IuliuHatieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania;
- Department of Surgery, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania
| |
Collapse
|
41
|
Tang B, Yang S. Involvement of Heparanase in Gastric Cancer Progression and Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:351-363. [PMID: 32274717 DOI: 10.1007/978-3-030-34521-1_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Heparanase is upregulated in various tumors, and its expression is closely associated with tumor growth, angiogenesis and metastasis, which accomplishes this mainly through degrading heparan sulfate and releasing heparin-binding growth factors thereby influencing multiple signaling pathways. In addition to its enzymatic degrading activity, heparanase can act via its non-enzymatic mechanisms that directly regulate various signaling. This review mainly focuses on the expression levels and role of heparanase in gastric cancer, and multiple genes and mechanisms regulating heparanase expression in gastric cancer. Furthermore, the development of heparanase-targeted immunotherapy and its potential application for treating gastric cancer are discussed.
Collapse
Affiliation(s)
- Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
42
|
Zhou Q, Tao X, Xia S, Guo F, Pan C, Xiang H, Shang D. T Lymphocytes: A Promising Immunotherapeutic Target for Pancreatitis and Pancreatic Cancer? Front Oncol 2020; 10:382. [PMID: 32266154 PMCID: PMC7105736 DOI: 10.3389/fonc.2020.00382] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/04/2020] [Indexed: 01/18/2023] Open
Abstract
Pancreatic disorders cause a broad spectrum of clinical diseases, mainly including acute and chronic pancreatitis and pancreatic cancer, and are associated with high global rates of morbidity and mortality. Unfortunately, the pathogenesis of pancreatic disease remains obscure, and there is a lack of specific treatments. T lymphocytes (T cells) play a vital role in the adaptive immune systems of multicellular organisms. During pancreatic disease development, local imbalances in T-cell subsets in inflammatory and tumor environments and the circulation have been observed. Furthermore, agents targeting T cells have been shown to reverse the natural course of pancreatic diseases. In this review, we have discussed the clinical relevance of T-cell alterations as a potential outcome predictor and the underlying mechanisms, as well as the present status of immunotherapy targeting T cells in pancreatitis and neoplasms. The breakthrough findings summarized in this review have important implications for innovative drug development and the prospective use of immunotherapy for pancreatitis and pancreatic cancer.
Collapse
Affiliation(s)
- Qi Zhou
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xufeng Tao
- School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Shilin Xia
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fangyue Guo
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Chen Pan
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of General Surgery, Pancreatic-Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hong Xiang
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- *Correspondence: Hong Xiang
| | - Dong Shang
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of General Surgery, Pancreatic-Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Dong Shang
| |
Collapse
|
43
|
Nichetti F, Marra A, Corti F, Guidi A, Raimondi A, Prinzi N, de Braud F, Pusceddu S. The Role of Mesothelin as a Diagnostic and Therapeutic Target in Pancreatic Ductal Adenocarcinoma: A Comprehensive Review. Target Oncol 2019; 13:333-351. [PMID: 29656320 DOI: 10.1007/s11523-018-0567-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesothelin is a tumor differentiation antigen, which is highly expressed in several solid neoplasms, including pancreatic cancer. Its selective expression on malignant cells and on only a limited number of healthy tissues has made it an interesting candidate for investigation as a diagnostic and prognostic biomarker and as a therapeutic target. Based on a strong preclinical rationale, a number of therapeutic agents targeting mesothelin have entered clinical trials, including immunotoxins, monoclonal antibodies, antibody-drug conjugates, cancer vaccines, and adoptive T cell therapies with chimeric antigen receptors. In pancreatic cancer, mesothelin has been investigated mainly to address two unmet issues: the urgent need for new laboratory techniques for early tumor detection and the lack of successfully targetable oncogenic alterations for patients' treatment. In this review, we describe the clinicopathological significance of mesothelin expression in pancreatic cancer initiation and progression, we summarize available studies evaluating mesothelin as a potential diagnostic and prognostic biomarker in this disease, and we discuss current evidence and future perspectives of preclinical and clinical studies testing mesothelin as a molecular target for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Federico Nichetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy.
| | - Antonio Marra
- Medical Oncology Unit, Azienda Ospedaliera San Paolo, Milan, Italy
| | - Francesca Corti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Alessandro Guidi
- Medical Oncology Unit, Azienda Ospedaliera San Gerardo, Monza, Italy
| | - Alessandra Raimondi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Natalie Prinzi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
- Department of Oncology, Università degli Studi di Milano, Milan, Italy
| | - Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| |
Collapse
|
44
|
Li T, Li H, Li S, Xu S, Zhang W, Gao H, Xu H, Wu C, Wang W, Yu X, Liu L. Research progress and design optimization of CAR-T therapy for pancreatic ductal adenocarcinoma. Cancer Med 2019; 8:5223-5231. [PMID: 31339230 PMCID: PMC6718528 DOI: 10.1002/cam4.2430] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/05/2019] [Accepted: 07/05/2019] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant cancer with limited treatment options. Chimeric antigen receptor T cells (CAR-T) are genetically engineered T cells that can specifically kill tumor cells without major histocompatibility complex restriction. Encouraging progress in CAR-T therapy for PDAC has been made in preclinical and early phase clinical trials. Challenges in CAR-T therapy for solid tumors still exist, including immunosuppressive microenvironment, interstitial barrier, poor chemotaxis, and the "on-target, off-tumor" effect. Applying neoantigens of PDAC as targets for CAR-T therapy, recognizing the CAR-T subgroup with better antitumor effect, and designing a CAR-T system targeting stroma of PDAC may contribute to develop a powerful CAR-T therapy for PDAC in the future.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm
- Biomarkers, Tumor
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/therapy
- Chemotaxis/immunology
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Research
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Microenvironment/immunology
- Pancreatic Neoplasms
Collapse
Affiliation(s)
- Tianjiao Li
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Hao Li
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Shuo Li
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Shuaishuai Xu
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Wuhu Zhang
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Heli Gao
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Huaxiang Xu
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Chuntao Wu
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Wenquan Wang
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Xianjun Yu
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Liang Liu
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| |
Collapse
|
45
|
Wang Z, Chen W, Zhang X, Cai Z, Huang W. A long way to the battlefront: CAR T cell therapy against solid cancers. J Cancer 2019; 10:3112-3123. [PMID: 31289581 PMCID: PMC6603378 DOI: 10.7150/jca.30406] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 04/07/2019] [Indexed: 12/15/2022] Open
Abstract
Chimeric antigen receptors (CARs) are engineered synthetic receptors that redirect and reprogram T cells to tumor surface antigens for subsequent eradication. The unprecedented efficacy of CD19-CAR T cells against B-cell malignancies has inspired oncologists to extend these efforts for the treatment of solid tumors. However, limited success has been achieved so far, partially due to some of the formidable challenges, e.g. suppression of full activation, inhibition of T cell localization, lacking of ideal targets, inefficient trafficking and infiltration, immunosuppression of microenvironment, and the probability of off targets and associated side effects. Significant progresses have being made recently. Thus, an updated summary is urgently needed. Here in this review, we discuss the advantages and some of the key hurdles encountered by CAR T cell therapy in solid tumors as well as the strategies adopted to improve therapeutic outcomes of this approach. Continuing efforts to increase therapeutic potential and decrease the adverse effects of adaptive cell transfer are suggested as well.
Collapse
Affiliation(s)
- Zhicai Wang
- Department of Medical Melanoma and Sarcoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518039, China
| | - Wei Chen
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518039, China
| | - Xing Zhang
- Department of Medical Melanoma and Sarcoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zhiming Cai
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518039, China.,Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen 518039, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen 518035, China
| | - Weiren Huang
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518039, China.,Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen 518039, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen 518035, China
| |
Collapse
|
46
|
Looi CK, Chung FFL, Leong CO, Wong SF, Rosli R, Mai CW. Therapeutic challenges and current immunomodulatory strategies in targeting the immunosuppressive pancreatic tumor microenvironment. J Exp Clin Cancer Res 2019; 38:162. [PMID: 30987642 PMCID: PMC6463646 DOI: 10.1186/s13046-019-1153-8] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/22/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Pancreatic cancer is one of the most lethal type of cancers, with an overall five-year survival rate of less than 5%. It is usually diagnosed at an advanced stage with limited therapeutic options. To date, no effective treatment options have demonstrated long-term benefits in advanced pancreatic cancer patients. Compared with other cancers, pancreatic cancer exhibits remarkable resistance to conventional therapy and possesses a highly immunosuppressive tumor microenvironment (TME). MAIN BODY In this review, we summarized the evidence and unique properties of TME in pancreatic cancer that may contribute to its resistance towards immunotherapies as well as strategies to overcome those barriers. We reviewed the current strategies and future perspectives of combination therapies that (1) promote T cell priming through tumor associated antigen presentation; (2) inhibit tumor immunosuppressive environment; and (3) break-down the desmoplastic barrier which improves tumor infiltrating lymphocytes entry into the TME. CONCLUSIONS It is imperative for clinicians and scientists to understand tumor immunology, identify novel biomarkers, and optimize the position of immunotherapy in therapeutic sequence, in order to improve pancreatic cancer clinical trial outcomes. Our collaborative efforts in targeting pancreatic TME will be the mainstay of achieving better clinical prognosis among pancreatic cancer patients. Ultimately, pancreatic cancer will be a treatable medical condition instead of a death sentence for a patient.
Collapse
Affiliation(s)
- Chin-King Looi
- 0000 0000 8946 5787grid.411729.8School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Felicia Fei-Lei Chung
- Mechanisms of Carcinogenesis Section (MCA), Epigenetics Group (EGE) International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Chee-Onn Leong
- 0000 0000 8946 5787grid.411729.8School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
- 0000 0000 8946 5787grid.411729.8Center for Cancer and Stem Cell Research, Institute for Research, Development and Innovation (IRDI), International Medical University, Kuala Lumpur, Malaysia
| | - Shew-Fung Wong
- 0000 0000 8946 5787grid.411729.8School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Rozita Rosli
- 0000 0001 2231 800Xgrid.11142.37UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Sri Kembangan, Selangor Malaysia
| | - Chun-Wai Mai
- 0000 0000 8946 5787grid.411729.8School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
- 0000 0000 8946 5787grid.411729.8Center for Cancer and Stem Cell Research, Institute for Research, Development and Innovation (IRDI), International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
47
|
Salvia R, Casciani F, Sereni E, Bassi C. Pancreatic cancer – What's next? Presse Med 2019; 48:e187-e197. [PMID: 30878338 DOI: 10.1016/j.lpm.2019.02.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/13/2019] [Indexed: 01/12/2023] Open
Abstract
This chapter focuses on the most recent advantages in the medical treatment of localized pancreatic cancer.
Collapse
Affiliation(s)
- Roberto Salvia
- University of Verona Hospital Trust, The Pancreas Institute, Unit of General and Pancreatic Surgery, Verona, Italy
| | - Fabio Casciani
- University of Verona Hospital Trust, The Pancreas Institute, Unit of General and Pancreatic Surgery, Verona, Italy.
| | - Elisabetta Sereni
- University of Verona Hospital Trust, The Pancreas Institute, Unit of General and Pancreatic Surgery, Verona, Italy
| | - Claudio Bassi
- University of Verona Hospital Trust, The Pancreas Institute, Unit of General and Pancreatic Surgery, Verona, Italy
| |
Collapse
|
48
|
Gene Therapy for Pancreatic Diseases: Current Status. Int J Mol Sci 2018; 19:ijms19113415. [PMID: 30384450 PMCID: PMC6275054 DOI: 10.3390/ijms19113415] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/22/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022] Open
Abstract
The pancreas is a key organ involved in digestion and endocrine functions in the body. The major diseases of the pancreas include pancreatitis, pancreatic cancer, cystic diseases, pancreatic divisum, islet cell tumors, endocrine tumors, diabetes mellitus, and pancreatic pain induced by these diseases. While various therapeutic methodologies have been established to date, however, the improvement of conventional treatments and establishment of novel therapies are essential to improve the efficacy. For example, conventional therapeutic options, including chemotherapy, are not effective against pancreatic cancer, and despite improvements in the last decade, the mortality rate has not declined and is estimated to become the second cause of cancer-related deaths by 2030. Therefore, continuous efforts focus on the development of novel therapeutic options. In this review, we will summarize the progress toward the development of gene therapies for pancreatic diseases, with an emphasis on recent preclinical studies and clinical trials. We aim to identify new areas for improvement of the current methodologies and new strategies that will lead to safe and effective gene therapeutic approaches in pancreatic diseases.
Collapse
|
49
|
DeSelm C, Palomba ML, Yahalom J, Hamieh M, Eyquem J, Rajasekhar VK, Sadelain M. Low-Dose Radiation Conditioning Enables CAR T Cells to Mitigate Antigen Escape. Mol Ther 2018; 26:2542-2552. [PMID: 30415658 DOI: 10.1016/j.ymthe.2018.09.008] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 09/07/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
CD19 chimeric antigen receptors (CARs) have demonstrated great efficacy against a range of B cell malignancies. However, antigen escape and, more generally, heterogeneous antigen expression pose a challenge to applying CAR therapy to a wide range of cancers. We find that low-dose radiation sensitizes tumor cells to immune rejection by locally activated CAR T cells. In a model of pancreatic adenocarcinoma heterogeneously expressing sialyl Lewis-A (sLeA), we show that not only sLeA+ but also sLeA- tumor cells exposed to low-dose radiation become susceptible to CAR therapy, reducing antigen-negative tumor relapse. RNA sequencing analysis of low-dose radiation-exposed tumors reveals the transcriptional signature of cells highly sensitive to TRAIL-mediated death. We find that sLeA-targeted CAR T cells produce TRAIL upon engaging sLeA+ tumor cells, and eliminate sLeA- tumor cells previously exposed to systemic or local low-dose radiation in a TRAIL-dependent manner. These findings enhance the prospects for successfully applying CAR therapy to heterogeneous solid tumors. Local radiation is integral to many tumors' standard of care and can be easily implemented as a CAR conditioning regimen.
Collapse
Affiliation(s)
- Carl DeSelm
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M Lia Palomba
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joachim Yahalom
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mohamad Hamieh
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Justin Eyquem
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Michel Sadelain
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Immunology Program, Sloan Kettering Institute, New York, NY, USA.
| |
Collapse
|
50
|
Pantuck M, Palaskas N, Drakaki A. Next generation T-cell therapy for genitourinary malignancies, part A: Introduction and current state of the art. Cancer Treat Res Commun 2018; 17:8-12. [PMID: 30173005 DOI: 10.1016/j.ctarc.2018.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/14/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Morgan Pantuck
- Department of Urology, Weill Cornell Medicine, United States.
| | - Nicolaos Palaskas
- Division of Hematology and Oncology, David Geffen School of Medicine, UCLA, Los Angeles, United States.
| | - Alexandra Drakaki
- Division of Hematology and Oncology, David Geffen School of Medicine, UCLA, Los Angeles, United States.
| |
Collapse
|