1
|
Huang H, Mu Y, Huang Y, Ji B, Wang Y, Chen CY, Chen Y, Luo Z, Li S, Zhang Z, Wang L, Conway JF, Yang D, Wang J, Sun J, Li S. Rational development of gemcitabine-based nanoplatform for targeting SERPINB9/Granzyme B axis to overcome chemo-immune-resistance. Nat Commun 2025; 16:4176. [PMID: 40325025 PMCID: PMC12053578 DOI: 10.1038/s41467-025-59490-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 04/25/2025] [Indexed: 05/07/2025] Open
Abstract
SERPINB9, an endogenous inhibitor of granzyme B (GzmB), has emerged as a critical factor in the resistance to immunotherapy by protecting cancer cells from GzmB-induced cytotoxicity. However, its role in chemosensitivity remains unknown. In this study, we show that gemcitabine (GEM) treatment upregulates SERPINB9 through transcription factor ATF-3. Interestingly, GEM also induces the expression of GzmB and knockout or knockdown of SERPINB9 results in enhanced response of tumor cells to GEM, suggesting a role of GzmB/SERPINB9 axis in regulating chemosensitivity. To facilitate the therapeutic translation of these findings, we engineer POEM nanocarrier (consisting of lipid-derivatized polylysine (PEG-PLL-Oleic acid, PPO), and GEM-conjugated polylysine (PEG-PLL-OA-GEM, PPOGEM), PPO/PPOGEM (POEM)) that is highly effective in codelivery of built-in GEM and loaded SERPINB9 short interfering RNA (siSPB9). GEM conjugation introduces an additional mechanism of carrier/siRNA interaction in addition to charge-mediated interaction and enables efficient i.v. delivery at lower N/P ratios. Here, we show that co-delivery of GEM and siSPB9 significantly improves antitumor efficacy and remodels the tumor immune microenvironment in pancreatic cancer models, supporting a promising therapeutic strategy.
Collapse
Affiliation(s)
- Haozhe Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yiqing Mu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yixian Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Beihong Ji
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yifei Wang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chien-Yu Chen
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yuang Chen
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhangyi Luo
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sihan Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ziqian Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Luxuan Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - James F Conway
- Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Da Yang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Junmei Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Jingjing Sun
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Yu J, Fu L, Wu R, Che L, Liu G, Ran Q, Xia Z, Liang X, Zhao G. Immunocytes in the tumor microenvironment: recent updates and interconnections. Front Immunol 2025; 16:1517959. [PMID: 40297580 PMCID: PMC12034658 DOI: 10.3389/fimmu.2025.1517959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
The tumor microenvironment (TME) is a complex, dynamic ecosystem where tumor cells interact with diverse immune and stromal cell types. This review provides an overview of the TME's evolving composition, emphasizing its transition from an early pro-inflammatory, immune-promoting state to a later immunosuppressive milieu characterized by metabolic reprogramming and hypoxia. It highlights the dual roles of key immunocytes-including T lymphocytes, natural killer cells, macrophages, dendritic cells, and myeloid-derived suppressor cells-which can either inhibit or support tumor progression based on their phenotypic polarization and local metabolic conditions. The article further elucidates mechanisms of immune cell plasticity, such as the M1/M2 macrophage switch and the balance between effector T cells and regulatory T cells, underscoring their impact on tumor growth and metastasis. Additionally, emerging therapeutic strategies, including checkpoint inhibitors and chimeric antigen receptor (CAR) T and NK cell therapies, as well as approaches targeting metabolic pathways, are discussed as promising avenues to reinvigorate antitumor immunity. By integrating recent molecular insights and clinical advancements, the review underscores the importance of deciphering the interplay between immunocytes and the TME to develop more effective cancer immunotherapies.
Collapse
Affiliation(s)
- Jiyao Yu
- Department of Ultrasound, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Li Fu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Gastroenterology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Rui Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Neurosurgery, Jiangyou People’s Hospital, Mianyang, China
| | - Linyi Che
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qinwen Ran
- General Practice Department, Wufu Town Hospital, Chongqing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China
| | - Xisong Liang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guanjian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Nemati M, Hsu CY, Nathiya D, Kumar MR, Oghenemaro EF, Kariem M, Kaur P, Bhanot D, Hjazi A, Azam Saedi T. Gemcitabine: immunomodulatory or immunosuppressive role in the tumor microenvironment. Front Immunol 2025; 16:1536428. [PMID: 40270972 PMCID: PMC12014622 DOI: 10.3389/fimmu.2025.1536428] [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: 11/28/2024] [Accepted: 03/21/2025] [Indexed: 04/25/2025] Open
Abstract
Gemcitabine (GEM), a nucleoside analog chemotherapy agent, has been widely used in the treatment of various cancers. In recent years, there has been growing interest in understanding the immunomodulatory or immunosuppressive effects of GEM. The immunomodulatory roles of GEM could influence the anti-tumor immune responses via several mechanisms, such as modulation of antigen presentation, cytokine production, and immune cell population. Furthermore, there is evidence that GEM enhances the therapeutic efficacy of immunotherapies, including oncolytic viruses, immune checkpoint inhibitors, CAR T-cells, and therapeutic vaccines. On the other hand, accumulating evidence also proposed that GEM may act as an immunosuppressive agent within the tumor microenvironment, resulting in immune evasion of tumor cells and tumor growth. These paradoxical roles of GEM in modifying immune responses highlight the complexity of GEM interaction with immune cells and responses within the tumor microenvironment. This review aims to provide an overview of the immunomodulatory and immunosuppressive effects of GEM within the tumor microenvironment and how GEM affects the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University, Phoenix, AZ, United States
| | - Deepak Nathiya
- Department of Pharmacy Practice, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - M. Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | - Deepak Bhanot
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Tayebeh Azam Saedi
- Department of Genetics, Faculty of Science, Islamic Azad University, Tonekabon, Iran
| |
Collapse
|
4
|
Tang J, Liu Z, Xie G, Wang C, Jiang Y. POU4F1 enhances lung cancer gemcitabine resistance by regulating METTL3-dependent TWF1 mRNA N6 adenosine methylation. 3 Biotech 2025; 15:7. [PMID: 39676891 PMCID: PMC11638459 DOI: 10.1007/s13205-024-04161-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/11/2024] [Indexed: 12/17/2024] Open
Abstract
This study aimed to investigate the role of POU Class 4 Homeobox 1 (POU4F1) in regulating gemcitabine (GEM) resistance in lung cancer cells. The mRNA and protein expressions were assessed using RT-qPCR, western blot, immunofluorescence, and immunohistochemistry. Cell viability and proliferation were assessed by CCK-8 assay and EdU assay. TUNEL staining and flow cytometry were employed to detect cell apoptosis. The m6A modification of TWF1 was detected using MeRIP assay. The interactions between molecules were validated using dual luciferase reporter gene, ChIP, and RIP assays. POU4F1 knockdown inhibited GEM resistance and autophagy in lung cancer cells. Mechanistically, POU4F1 transcriptionally activated methyltransferase-like protein 3 (METTL3) in GEM-resistant cells by binding to the METTL3 promoter. METTL3 promoted the N6-methyladenosine (m6A) modification and expression level of twinfilin-1 (TWF1). Overexpression of METTL3 and TWF1 weakened the effects of POU4F1 knockdown on GEM resistance and autophagy. Moreover, knockdown POU4F1 also enhanced GEM anti-tumor sensitivity in vivo. In conclusion, POU4F1 upregulation promoted GEM resistance in lung cancer cells by promoting autophagy through increasing METTL3-mediated TWF1 m6A modification. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-024-04161-w.
Collapse
Affiliation(s)
- Jianfeng Tang
- Department of Cardiovascular Thoracic Surgery, The Central Hospital of Yongzhou, No. 396 Yiyun Road, Lengshuitan District, Yongzhou, 425100 Hunan People’s Republic of China
| | - Zhijian Liu
- Department of Cardiovascular Thoracic Surgery, The Central Hospital of Yongzhou, No. 396 Yiyun Road, Lengshuitan District, Yongzhou, 425100 Hunan People’s Republic of China
| | - Guanghui Xie
- Department of Cardiovascular Thoracic Surgery, The Central Hospital of Yongzhou, No. 396 Yiyun Road, Lengshuitan District, Yongzhou, 425100 Hunan People’s Republic of China
| | - Chenbin Wang
- Department of Cardiovascular Thoracic Surgery, The Central Hospital of Yongzhou, No. 396 Yiyun Road, Lengshuitan District, Yongzhou, 425100 Hunan People’s Republic of China
| | - Yongjun Jiang
- Department of Cardiovascular Thoracic Surgery, The Central Hospital of Yongzhou, No. 396 Yiyun Road, Lengshuitan District, Yongzhou, 425100 Hunan People’s Republic of China
| |
Collapse
|
5
|
Chen S, Zhu H, Jounaidi Y. Comprehensive snapshots of natural killer cells functions, signaling, molecular mechanisms and clinical utilization. Signal Transduct Target Ther 2024; 9:302. [PMID: 39511139 PMCID: PMC11544004 DOI: 10.1038/s41392-024-02005-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 11/15/2024] Open
Abstract
Natural killer (NK) cells, initially identified for their rapid virus-infected and leukemia cell killing and tumor destruction, are pivotal in immunity. They exhibit multifaceted roles in cancer, viral infections, autoimmunity, pregnancy, wound healing, and more. Derived from a common lymphoid progenitor, they lack CD3, B-cell, or T-cell receptors but wield high cytotoxicity via perforin and granzymes. NK cells orchestrate immune responses, secreting inflammatory IFNγ or immunosuppressive TGFβ and IL-10. CD56dim and CD56bright NK cells execute cytotoxicity, while CD56bright cells also regulate immunity. However, beyond the CD56 dichotomy, detailed phenotypic diversity reveals many functional subsets that may not be optimal for cancer immunotherapy. In this review, we provide comprehensive and detailed snapshots of NK cells' functions and states of activation and inhibitions in cancer, autoimmunity, angiogenesis, wound healing, pregnancy and fertility, aging, and senescence mediated by complex signaling and ligand-receptor interactions, including the impact of the environment. As the use of engineered NK cells for cancer immunotherapy accelerates, often in the footsteps of T-cell-derived engineering, we examine the interactions of NK cells with other immune effectors and relevant signaling and the limitations in the tumor microenvironment, intending to understand how to enhance their cytolytic activities specifically for cancer immunotherapy.
Collapse
Affiliation(s)
- Sumei Chen
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China.
| | - Haitao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Youssef Jounaidi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Khaliulin M, Valiullina A, Petukhov A, Yuan Y, Spada S, Bulatov E. Breaking the shield of solid tumors: a combined approach for enhanced efficacy of CAR-T cells. Cancer Immunol Immunother 2024; 74:3. [PMID: 39487875 PMCID: PMC11531461 DOI: 10.1007/s00262-024-03817-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 08/22/2024] [Indexed: 11/04/2024]
Abstract
The use of chimeric antigen receptor (CAR)-T cells has enhanced the range of available therapeutic modalities in the context of cancer treatment. CAR-T cells have demonstrated considerable efficacy in the targeted eradication of blood cancer cells, thereby stimulating substantial interest in the advancement of such therapeutic approaches. However, the efficacy of CAR-T cells against solid tumor cells has been limited due to the presence of various obstacles. Solid tumors exhibit antigenic diversity and an immunosuppressive microenvironment, which presents a challenge for immune cells attempting to penetrate the tumor. CAR-T cells also demonstrate decreased proliferative activity and cytotoxicity. Furthermore, concerns exist regarding tumor antigen loss and therapy-associated toxicity. Currently, scientists are working to enhance the structure of the CAR and improve the survival and efficiency of CAR-T cells in recognizing tumor antigens in solid tumors. Chemotherapy drugs are frequently employed in the treatment of malignant neoplasms and can also be used prior to cell therapy to enhance CAR-T cell engraftment. Recent studies have demonstrated that chemotherapy drugs can mitigate the suppressive impact of TME, eliminate the physical barrier by destroying the tumor stroma, and facilitate greater penetration of immune cells and CAR-T cells into the tumor. This, in turn, increases their survival, persistence, and cytotoxicity, as well as affects the metabolism of immune cells inside the tumor. However, the effectiveness of the combined approach against solid tumors depends on several factors, including the type of tumor, dosage, population of CAR-T cells, and individual characteristics of the body. This review examines the principal obstacles to the utilization of CAR-T cells against solid tumors, proposes solutions to these issues, and assesses the potential advantages of a combined approach to radiation exposure, which has the potential to enhance the sensitivity of the tumor to other agents.
Collapse
Affiliation(s)
- Marat Khaliulin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia, 420008
| | - Aygul Valiullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia, 420008
| | - Alexey Petukhov
- Nazarbaev University, Qabanbay Batyr Ave 53, 010000, Astana, Kazakhstan
| | - Youyong Yuan
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, People's Republic of China
| | - Sheila Spada
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Emil Bulatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia, 420008.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia, 117997.
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia, 119048.
| |
Collapse
|
7
|
Metselaar DS, Meel MH, Goulding JR, du Chatinier A, Rigamonti L, Waranecki P, Geisemeyer N, de Gooijer MC, Breur M, Koster J, Veldhuijzen van Zanten SEM, Bugiani M, Franke NE, Reddy A, Wesseling P, Kaspers GJL, Hulleman E. Gemcitabine therapeutically disrupts essential SIRT1-mediated p53 repression in atypical teratoid/rhabdoid tumors. Cell Rep Med 2024; 5:101700. [PMID: 39208799 PMCID: PMC11524974 DOI: 10.1016/j.xcrm.2024.101700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/13/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024]
Abstract
Atypical teratoid/rhabdoid tumors (ATRTs) are highly malignant embryonal tumors of the central nervous system with a dismal prognosis. Using a newly developed and validated patient-derived ATRT culture and xenograft model, alongside a panel of primary ATRT models, we found that ATRTs are selectively sensitive to the nucleoside analog gemcitabine. Gene expression and protein analyses indicate that gemcitabine treatment causes the degradation of sirtuin 1 (SIRT1), resulting in cell death through activation of nuclear factor κB (NF-κB) and p53. Furthermore, we discovered that gemcitabine-induced loss of SIRT1 results in a nucleus-to-cytoplasm translocation of the sonic hedgehog (SHH) signaling activator GLI2, explaining the observed additional gemcitabine sensitivity in SHH-subtype ATRT. Treatment of ATRT xenograft-bearing mice with gemcitabine resulted in a >30% increase in median survival and yielded long-term survivors in two independent patient-derived xenograft models. These findings demonstrate that ATRTs are highly sensitive to gemcitabine treatment and may form part of a future multimodal treatment strategy for ATRTs.
Collapse
Affiliation(s)
- Dennis S Metselaar
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Michaël H Meel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Joshua R Goulding
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | | | - Leyla Rigamonti
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Piotr Waranecki
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Neal Geisemeyer
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; Division of Pediatric Neuro-Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Mark C de Gooijer
- Division of Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marjolein Breur
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Jan Koster
- Department of Epidemiology and Biostatistics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | | | - Marianna Bugiani
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Niels E Franke
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Alyssa Reddy
- Departments of Neurology and Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Pieter Wesseling
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pathology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Gertjan J L Kaspers
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Scolaro T, Manco M, Pecqueux M, Amorim R, Trotta R, Van Acker HH, Van Haele M, Shirgaonkar N, Naulaerts S, Daniluk J, Prenen F, Varamo C, Ponti D, Doglioni G, Ferreira Campos AM, Fernandez Garcia J, Radenkovic S, Rouhi P, Beatovic A, Wang L, Wang Y, Tzoumpa A, Antoranz A, Sargsian A, Di Matteo M, Berardi E, Goveia J, Ghesquière B, Roskams T, Soenen S, Voets T, Manshian B, Fendt SM, Carmeliet P, Garg AD, DasGupta R, Topal B, Mazzone M. Nucleotide metabolism in cancer cells fuels a UDP-driven macrophage cross-talk, promoting immunosuppression and immunotherapy resistance. NATURE CANCER 2024; 5:1206-1226. [PMID: 38844817 PMCID: PMC11358017 DOI: 10.1038/s43018-024-00771-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/23/2024] [Indexed: 08/16/2024]
Abstract
Many individuals with cancer are resistant to immunotherapies. Here, we identify the gene encoding the pyrimidine salvage pathway enzyme cytidine deaminase (CDA) among the top upregulated metabolic genes in several immunotherapy-resistant tumors. We show that CDA in cancer cells contributes to the uridine diphosphate (UDP) pool. Extracellular UDP hijacks immunosuppressive tumor-associated macrophages (TAMs) through its receptor P2Y6. Pharmacologic or genetic inhibition of CDA in cancer cells (or P2Y6 in TAMs) disrupts TAM-mediated immunosuppression, promoting cytotoxic T cell entry and susceptibility to anti-programmed cell death protein 1 (anti-PD-1) treatment in resistant pancreatic ductal adenocarcinoma (PDAC) and melanoma models. Conversely, CDA overexpression in CDA-depleted PDACs or anti-PD-1-responsive colorectal tumors or systemic UDP administration (re)establishes resistance. In individuals with PDAC, high CDA levels in cancer cells correlate with increased TAMs, lower cytotoxic T cells and possibly anti-PD-1 resistance. In a pan-cancer single-cell atlas, CDAhigh cancer cells match with T cell cytotoxicity dysfunction and P2RY6high TAMs. Overall, we suggest CDA and P2Y6 as potential targets for cancer immunotherapy.
Collapse
Affiliation(s)
- Tommaso Scolaro
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Marta Manco
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Mathieu Pecqueux
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ricardo Amorim
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rosa Trotta
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Heleen H Van Acker
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Matthias Van Haele
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Niranjan Shirgaonkar
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Stefan Naulaerts
- Laboratory for Cell Stress & Immunity (CSI), Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jan Daniluk
- Laboratory of Ion Channel Research (LICR), VIB-KU Leuven Centre for Brain & Disease Research, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Fran Prenen
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Chiara Varamo
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Donatella Ponti
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Latina, Italy
| | - Ginevra Doglioni
- Laboratory of Cellular Metabolism and Metabolic Regulation, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Ana Margarida Ferreira Campos
- Laboratory of Cellular Metabolism and Metabolic Regulation, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Juan Fernandez Garcia
- Laboratory of Cellular Metabolism and Metabolic Regulation, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Silvia Radenkovic
- Metabolomics Core Facility, Center for Cancer Biology, VIB, Leuven, Belgium
- Metabolomics Core Facility, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Pegah Rouhi
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Liwei Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Amalia Tzoumpa
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Asier Antoranz
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Ara Sargsian
- Translation Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Mario Di Matteo
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Emanuele Berardi
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jermaine Goveia
- Unicle Biomedical Data Science, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Bart Ghesquière
- Metabolomics Core Facility, Center for Cancer Biology, VIB, Leuven, Belgium
- Metabolomics Core Facility, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Tania Roskams
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Stefaan Soenen
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research (LICR), VIB-KU Leuven Centre for Brain & Disease Research, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Bella Manshian
- Translation Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Laboratory for Cell Stress & Immunity (CSI), Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ramanuj DasGupta
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Baki Topal
- Department of Visceral Surgery, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium.
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
9
|
Santucci KL, Snyder KK, Van Buskirk RG, Baust JG, Baust JM. Investigation of Lung Cancer Cell Response to Cryoablation and Adjunctive Gemcitabine-Based Cryo-Chemotherapy Using the A549 Cell Line. Biomedicines 2024; 12:1239. [PMID: 38927445 PMCID: PMC11200978 DOI: 10.3390/biomedicines12061239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/17/2024] [Accepted: 05/25/2024] [Indexed: 06/28/2024] Open
Abstract
Due to the rising annual incidence of lung cancer (LC), new treatment strategies are needed. While various options exist, many, if not all, remain suboptimal. Several studies have shown cryoablation to be a promising approach. Yet, a lack of basic information pertaining to LC response to freezing and requirement for percutaneous access has limited clinical use. In this study, we investigated the A549 lung carcinoma cell line response to freezing. The data show that a single 5 min freeze to -15 °C did not affect cell viability, whereas -20 °C and -25 °C result in a significant reduction in viability 1 day post freeze to <10%. These populations, however, were able to recover in culture. Application of a repeat (double) freeze resulted in complete cell death at -25 °C. Studies investigating the impact of adjunctive gemcitabine (75 nM) pretreatment in combination with freezing were then conducted. Exposure to gemcitabine alone resulted in minimal cell death. The combination of gemcitabine pretreatment and a -20 °C single freeze as well as combination treatment with a -15 °C repeat freeze both resulted in complete cell death. This suggests that gemcitabine pretreatment may be synergistically effective when combined with freezing. Studies into the modes of cell death associated with the increased cell death revealed the increased involvement of necroptosis in combination treatment. In summary, these results suggest that repeat freezing to -20 °C to -25 °C results in a high degree of LC destruction. Further, the data suggest that the combination of gemcitabine pretreatment and freezing resulted in a shift of the minimum lethal temperature for LC from -25 °C to -15 °C. These findings, in combination with previous reports, suggest that cryoablation alone or in combination with chemotherapy may provide an improved path for the treatment of LC.
Collapse
Affiliation(s)
| | - Kristi K. Snyder
- CPSI Biotech, Owego, NY 13827, USA
- Phase Therapeutics, Inc., Owego, NY 13827, USA
| | - Robert G. Van Buskirk
- CPSI Biotech, Owego, NY 13827, USA
- Center for Translational Stem Cell and Tissue Engineering, Binghamton University, Binghamton, NY 13902, USA
- Department of Biological Sciences, Binghamton University, Binghamton, NY 13902, USA
| | - John G. Baust
- Center for Translational Stem Cell and Tissue Engineering, Binghamton University, Binghamton, NY 13902, USA
- Department of Biological Sciences, Binghamton University, Binghamton, NY 13902, USA
| | - John M. Baust
- CPSI Biotech, Owego, NY 13827, USA
- Phase Therapeutics, Inc., Owego, NY 13827, USA
| |
Collapse
|
10
|
Salim EI, Elsebakhy S, Hessien M. Repurposing of atorvastatin and metformin denotes their individual and combined antiproliferative effects in non-small cell lung cancer. Fundam Clin Pharmacol 2024; 38:550-560. [PMID: 38258539 DOI: 10.1111/fcp.12981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 12/11/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024]
Abstract
BACKGROUND Due to the limited success in the treatment of lung adenocarcinomas, new treatment protocols are urgently needed to increase the curability rate and the survival of lung cancer patients. OBJECTIVES Although statins, like atorvastatin (Ator), and metformin (Met) are widely accepted as hypolipidemic and hypoglycemic drugs, respectively, there are many predictions about their enhancing antitumor effect when they are combined with traditional chemotherapeutics. METHODS The individual and combined antiproliferative potential of Ator and Met was tested by MTT-assay in non-small cell lung cancer (NSCLC) A549 cell line, compared to the corresponding effect of Gemcitabine (Gem) with implication on the mechanisms of action. RESULTS Initially, both drugs demonstrated concentration-dependent cytotoxicity in A549 cells. Also, their combination index (CI) indicated their synergistic effect at equi-IC50 concentration (CI = 0.00984). Moreover, Ator and/or Met-treated cells revealed disrupted patterns of SOD, CAT, GSH, MDA, and TAC, developed apoptosis, and larger fractions of the cell population were arrested in G0/G1 phase, particularly in cells dually-treated both Ator and Met. These observations were accompanied by downregulation in the expression of iNOS, HO-1, and the angiogenic marker VEGF, meanwhile, an altered expression of MAPK and AMPK was observed. CONCLUSION Conclusively, these data suggest that repurposing of Ator and Met demonstrates their individual and combined antiproliferative effect in non-small cell lung cancer and they may adopt a similar mechanism of action.
Collapse
Affiliation(s)
- Elsayed I Salim
- Zoology Department, Research Lab. of Molecular Carcinogenesis, Faculty of Science, Tanta University, Tanta, Egypt
| | - Safaa Elsebakhy
- Molecular Cell Biology Unit, Division of Biochemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Mohamed Hessien
- Molecular Cell Biology Unit, Division of Biochemistry, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
11
|
Zhang SL, Tian Y, Yu J, Zhang JH, Sun L, Huang LT, Ma JT, Han CB. Is neoadjuvant immunotherapy necessary in patients with programmed death ligand 1 expression-negative resectable non-small cell lung cancer? A systematic review and meta-analysis. Lung Cancer 2024; 191:107799. [PMID: 38669725 DOI: 10.1016/j.lungcan.2024.107799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/30/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024]
Abstract
OBJECTIVES The aim of this study was to investigate the clinical benefit and necessity of neoadjuvant programmed cell death (or ligand) (PD-(L)1) blockades in resectable non-small cell lung cancer (NSCLC) patients with negative PD-L1 expression. MATERIALS AND METHODS Randomized control trials (RCTs) that compared event-free survival (EFS), overall survival (OS), major pathological response (MPR), and/or pathological complete response (pCR) between neoadjuvant chemo-immunotherapy (nCIT) and neoadjuvant chemotherapy (nCT) for patients with resectable NSCLC stratified by PD-L1 expression were eligible for inclusion in the study. Data regarding the pathological response and EFS were evaluated by the odds ratio (OR) and hazard ratio (HR) with 95% confidence interval (CI) using random and fixed models. RESULTS A total of six RCTs involving 3,194 patients with resectable NSCLC with or without neoadjuvant immunotherapy were included. Compared with nCT alone, nCIT significantly improved pCR (18.3 % vs. 3.0 %; OR, 5.64; 95 % CI, 3.22-9.89; P < 0.001), MPR (38.9 % vs. 15.5 %; OR, 3.57; 95 % CI, 2.10-6.05; P < 0.001), and EFS (HR, 0.75; 95 % CI, 0.62-0.90; P = 0.002) in PD-L1 <1 % NSCLC patients. In addition, PD-L1 ≥1 % was associated with higher rates of pCR (32.8 % vs. 18.3 %; OR, 2.28; 95 % CI, 1.40-3.73; P = 0.001) and MPR (53.9 % vs. 38.9 %; OR, 1.84; 95 % CI, 1.22-2.79; P = 004) and longer EFS (HR, 0.44 vs. 0.75) in the setting of nCIT compared with PD-L1 <1 %. nCIT improved only OS in NSCLC patients with PD-L1 ≥1 % but not in patients with PD-L1 <1 %. CONCLUSIONS The use of nCIT should be recommended for resectable NSCLC patients with negative PD-L1 expression, as nCIT significantly improved the pathological response and EFS in these patients. The benefit to PD-L1-negative patients treated with nCIT on OS remains to be validated.
Collapse
Affiliation(s)
- Shu-Ling Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Yuan Tian
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Jing Yu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Jie-Hui Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Li Sun
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Le-Tian Huang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Jie-Tao Ma
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Cheng-Bo Han
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
12
|
Larson AC, Doty KR, Solheim JC. The double life of a chemotherapy drug: Immunomodulatory functions of gemcitabine in cancer. Cancer Med 2024; 13:e7287. [PMID: 38770637 PMCID: PMC11106691 DOI: 10.1002/cam4.7287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 05/22/2024] Open
Abstract
Although the development of immunotherapies has been revolutionary in the treatment of several cancers, many cancer types remain unresponsive to immune-based treatment and are largely managed by chemotherapy drugs. However, chemotherapeutics are not infallible and are frequently rendered ineffective as resistance develops from prolonged exposure. Recent investigations have indicated that some chemotherapy drugs have additional functions beyond their normative cytotoxic capacity and are in fact immune-modifying agents. Of the pharmaceuticals with identified immune-editing properties, gemcitabine is well-studied and of interest to clinicians and scientists alike. Gemcitabine is a chemotherapy drug approved for the treatment of multiple cancers, including breast, lung, pancreatic, and ovarian. Because of its broad applications, relatively low toxicity profile, and history as a favorable combinatory partner, there is promise in the recharacterization of gemcitabine in the context of the immune system. Such efforts may allow the identification of suitable immunotherapeutic combinations, wherein gemcitabine can be used as a priming agent to improve immunotherapy efficacy in traditionally insensitive cancers. This review looks to highlight documented immunomodulatory abilities of one of the most well-known chemotherapy agents, gemcitabine, relating to its influence on cells and proteins of the immune system.
Collapse
Affiliation(s)
- Alaina C. Larson
- Eppley Institute for Research in Cancer & Allied DiseasesUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Kenadie R. Doty
- Eppley Institute for Research in Cancer & Allied DiseasesUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Joyce C. Solheim
- Eppley Institute for Research in Cancer & Allied DiseasesUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Department of Biochemistry & Molecular BiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Department of Pathology, Microbiology, & ImmunologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
13
|
Liu J, Han Y, Zhao M, Wang L, Hu H, Chen D. Unlocking the power of immunotherapy: Combinatorial delivery of plasmid IL-15 and gemcitabine to synergistically remodeling the tumor microenvironment. Int J Pharm 2024; 655:124027. [PMID: 38554742 DOI: 10.1016/j.ijpharm.2024.124027] [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: 11/30/2023] [Revised: 03/03/2024] [Accepted: 03/19/2024] [Indexed: 04/02/2024]
Abstract
Cancer immunotherapy has emerged as a promising clinical treatment strategy in recent years. Unfortunately, the satisfactory antitumor therapeutic efficacy of immunotherapy is limited by intricate immunosuppressive tumor microenvironment (ITM). To remodel the ITM and alleviate the immune evasion, we constructed FA-PEG-modified liposomes to deliver plasmid IL-15 (pIL-15) and gemcitabine (GEM) (FPCL@pIL-15 + FPGL), respectively. The FPCL@pIL-15 (150 nm) and FPGL (120 nm) exhibited symmetrically spherical structures as well as desirable penetration and accumulation on tumor tissue depending on folic acid (FA) specialized targeting function. The transfected expression of IL-15 efficiently fosters the proliferation and co-activation of Natural killer (NK) cells and CD8+T cells through binding to IL-15R. FPGL upregulated the expression of Natural killer group 2 member D ligands (NKG2DLs) and reinforced recognition by NK cells to alleviate the immune evasion, and simultaneously promoted activation of CD8+T cells through immunogenic cell death (ICD) effects. More importantly, the combinatorial administration achieved intended anti-tumor efficacy in the subcutaneous 4T1 tumor model. In essence, we demonstrated that combining FPCL@pIL-15 with FPGL synergistically stimulates and mobilizes the immune system to reverse the ITM and trigger an anti-tumor immune response, indicating a tremendous potential for application in immunotherapy.
Collapse
Affiliation(s)
- Jingwen Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Yanyan Han
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Ming Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Leyuan Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China
| | - Haiyang Hu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China.
| | - Dawei Chen
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, PR China.
| |
Collapse
|
14
|
Khaliulin MR, Safin RN, Kunst MA, Bulatov ER. The use of T-cells with chimeric antigen receptor (CAR-T) in combination with chemotherapy and radiotherapy for the treatment of solid tumors. ADVANCES IN MOLECULAR ONCOLOGY 2024; 11:31-45. [DOI: 10.17650/2313-805x-2024-11-1-31-45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The introduction of chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of hematological diseases, particularly in combating blood cancer. The success of this cell therapy approach has led to the development of approximately seven commercial CAR-T based drugs. However, the application of CAR-T therapy for solid tumors has proven to be less effective due to challenges such as the varied antigens in solid tumors, an immunosuppressive tumor environment, limited immune cell infiltration, reduced CAR-T cell activity and toxicity issues. To solve these problems, scientists are making efforts to improve and improve the methods of treatment of solid tumors. Chemotherapy is the standard treatment for a large number of malignant neoplasms. It is also used before starting cell therapy for lymphodepletion and better engraftment of injected CAR-T cells. It has been shown that chemotherapy can reduce the immunosuppressive effect of the tumor microenvironment, destroy the stroma, and promote better infiltration of the tumor by CAR-T cells, improving their survival, persistence, cytotoxicity, and influencing the metabolism of immune cells inside the tumor. The effectiveness of combining chemotherapy and CAR-T cell therapy relies on various factors such as tumor type, dosage, treatment schedule, CAR-T cell composition, and individual biological traits. Similarly, radiation therapy can enhance tumor cell vulnerability to specific treatments while also supporting tumor cell survival.In this review, we discuss the use of CAR-T therapy to combat solid tumors, regarding the challenges of treating solid tumors, ways to overcome them, and also touch upon the possibility of using combination treatments to improve the effectiveness of cell therapy.
Collapse
Affiliation(s)
| | - R. N. Safin
- Republican Clinical Oncology Dispensary named after Prof. M.Z. Sigal Russia
| | - M. A. Kunst
- Republican Clinical Hospital of the Ministry of Health of the Republic of Tatarstan
| | - E. R. Bulatov
- Kazan (Volga Region) Federal University; Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences
| |
Collapse
|
15
|
Larson AC, Knoche SM, Brumfield GL, Doty KR, Gephart BD, Moore-Saufley PR, Solheim JC. Gemcitabine Modulates HLA-I Regulation to Improve Tumor Antigen Presentation by Pancreatic Cancer Cells. Int J Mol Sci 2024; 25:3211. [PMID: 38542184 PMCID: PMC10970070 DOI: 10.3390/ijms25063211] [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: 01/30/2024] [Revised: 02/25/2024] [Accepted: 03/04/2024] [Indexed: 04/26/2024] Open
Abstract
Pancreatic cancer is a lethal disease, harboring a five-year overall survival rate of only 13%. Current treatment approaches thus require modulation, with attention shifting towards liberating the stalled efficacy of immunotherapies. Select chemotherapy drugs which possess inherent immune-modifying behaviors could revitalize immune activity against pancreatic tumors and potentiate immunotherapeutic success. In this study, we characterized the influence of gemcitabine, a chemotherapy drug approved for the treatment of pancreatic cancer, on tumor antigen presentation by human leukocyte antigen class I (HLA-I). Gemcitabine increased pancreatic cancer cells' HLA-I mRNA transcripts, total protein, surface expression, and surface stability. Temperature-dependent assay results indicated that the increased HLA-I stability may be due to reduced binding of low affinity peptides. Mass spectrometry analysis confirmed changes in the HLA-I-presented peptide pool post-treatment, and computational predictions suggested improved affinity and immunogenicity of peptides displayed solely by gemcitabine-treated cells. Most of the gemcitabine-exclusive peptides were derived from unique source proteins, with a notable overrepresentation of translation-related proteins. Gemcitabine also increased expression of select immunoproteasome subunits, providing a plausible mechanism for its modulation of the HLA-I-bound peptidome. Our work supports continued investigation of immunotherapies, including peptide-based vaccines, to be used with gemcitabine as new combination treatment modalities for pancreatic cancer.
Collapse
Affiliation(s)
- Alaina C. Larson
- Eppley Institute for Research in Cancer & Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Shelby M. Knoche
- Eppley Institute for Research in Cancer & Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Gabrielle L. Brumfield
- Eppley Institute for Research in Cancer & Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kenadie R. Doty
- Eppley Institute for Research in Cancer & Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Benjamin D. Gephart
- Eppley Institute for Research in Cancer & Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | - Joyce C. Solheim
- Eppley Institute for Research in Cancer & Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
16
|
Huang X, Ren Q, Yang L, Cui D, Ma C, Zheng Y, Wu J. Immunogenic chemotherapy: great potential for improving response rates. Front Oncol 2023; 13:1308681. [PMID: 38125944 PMCID: PMC10732354 DOI: 10.3389/fonc.2023.1308681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
The activation of anti-tumor immunity is critical in treating cancers. Recent studies indicate that several chemotherapy agents can stimulate anti-tumor immunity by inducing immunogenic cell death and durably eradicate tumors. This suggests that immunogenic chemotherapy holds great potential for improving response rates. However, chemotherapy in practice has only had limited success in inducing long-term survival or cure of cancers when used either alone or in combination with immunotherapy. We think that this is because the importance of dose, schedule, and tumor model dependence of chemotherapy-activated anti-tumor immunity is under-appreciated. Here, we review immune modulation function of representative chemotherapy agents and propose a model of immunogenic chemotherapy-induced long-lasting responses that rely on synergetic interaction between killing tumor cells and inducing anti-tumor immunity. We comb through several chemotherapy treatment schedules, and identify the needs for chemotherapy dose and schedule optimization and combination therapy with immunotherapy when chemotherapy dosage or immune responsiveness is too low. We further review tumor cell intrinsic factors that affect the optimal chemotherapy dose and schedule. Lastly, we review the biomarkers indicating responsiveness to chemotherapy and/or immunotherapy treatments. A deep understanding of how chemotherapy activates anti-tumor immunity and how to monitor its responsiveness can lead to the development of more effective chemotherapy or chemo-immunotherapy, thereby improving the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Xiaojun Huang
- Cancer Center, Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qinghuan Ren
- Alberta Institute, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Leixiang Yang
- Cancer Center, The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Center for Reproductive Medicine, Department of Genetic and Genomic Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Di Cui
- Cancer Center, The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chenyang Ma
- Department of Internal Medicine of Traditional Chinese Medicine, The Second People’s Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Yueliang Zheng
- Cancer Center, Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Junjie Wu
- Cancer Center, The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Center for Reproductive Medicine, Department of Genetic and Genomic Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
17
|
Fu T, Ma X, Du SL, Ke ZY, Wang XC, Yin HH, Wang WX, Liu YJ, Liang AL. p21 promotes gemcitabine tolerance in A549 cells by inhibiting DNA damage and altering the cell cycle. Oncol Lett 2023; 26:471. [PMID: 37809050 PMCID: PMC10551858 DOI: 10.3892/ol.2023.14059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
Gemcitabine is one of the most widely used chemotherapy drugs for advanced malignant tumors, including non-small cell lung cancer. However, the clinical efficacy of gemcitabine is limited due to drug resistance. The aim of the present study was to investigate the role of p21 in gemcitabine-resistant A549 (A549/G+) lung cancer cells. IC50 values were determined using a Cell Counting Kit-8 (CCK-8) assay. mRNA and protein expression levels of genes were measured by reverse transcription-quantitative PCR and western blotting, respectively. The cell cycle distribution and apoptosis rate were analyzed by flow cytometry. DNA damage in cells was evaluated by single-cell gel electrophoresis. The results of western blot analysis and the CCK-8 assay demonstrated that the expression of p21 was higher in A549/G+ cells than in gemcitabine-sensitive cells. Knockdown of p21 expression in gemcitabine-resistant cells sensitized these cells to gemcitabine (with the IC50 decreasing from 84.2 to 26.7 µM). Cell cycle analysis revealed different changes in the cell cycle distribution in A549/G+ cells treated with the same concentration of gemcitabine, and decreased expression of p21 was shown to promote G1 arrest. The apoptosis assay and comet assay results revealed that decreased p21 expression resulted in accumulation of unrepaired DNA double-strand breaks (DSBs) and induction of apoptosis by gemcitabine. The present study demonstrated that knockout of p21 mRNA expression in A549/G+ cells promotes apoptosis and DNA DSB accumulation, accompanied by G1 arrest. These results indicated that p21 is involved in regulating the response of A549 cells to gemcitabine.
Collapse
Affiliation(s)
- Tian Fu
- Department of Biochemistry and Molecular Biology, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
- Department of Clinical Biochemistry, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
- Department of Clinical Laboratory, Zhanjiang Central Hospital, Zhanjiang, Guangdong 524045, P.R. China
| | - Xuan Ma
- Department of Clinical Laboratory, Xinle City Hospital, Shijiazhuang, Hebei 050700, P.R. China
| | - Shen-Lin Du
- Department of Clinical Laboratory, Dongguan People's Hospital, Dongguan, Guangdong 523058, P.R. China
| | - Zhi-Yin Ke
- Department of Biochemistry and Molecular Biology, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
- Department of Clinical Biochemistry, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Xue-Chun Wang
- Department of Biochemistry and Molecular Biology, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
- Department of Clinical Biochemistry, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Hai-Han Yin
- Department of Biochemistry and Molecular Biology, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
- Department of Clinical Biochemistry, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Wen-Xuan Wang
- Department of Biochemistry and Molecular Biology, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
- Department of Clinical Biochemistry, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Yong-Jun Liu
- Department of Biochemistry and Molecular Biology, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
- Department of Clinical Biochemistry, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Ai-Ling Liang
- Department of Biochemistry and Molecular Biology, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
- Department of Clinical Biochemistry, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| |
Collapse
|
18
|
Chen X, Tao Z, Liang Y, Ma M, Adah D, Ding W, Chen L, Li X, Dai L, Fanuel S, Zhao S, Hu W, Wu D, Duan Z, Zhou F, Qin L, Chen X, Yang Z. Plasmodium immunotherapy combined with gemcitabine has a synergistic inhibitory effect on tumor growth and metastasis in murine Lewis lung cancer models. Front Oncol 2023; 13:1181176. [PMID: 37916167 PMCID: PMC10618005 DOI: 10.3389/fonc.2023.1181176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 09/18/2023] [Indexed: 11/03/2023] Open
Abstract
Objective Our previous studies have demonstrated that Plasmodium immunotherapy (infection) has antitumor effects in mice. However, as a new form of immunotherapy, this therapy has a weakness: its specific killing effect on tumor cells is relatively weak. Therefore, we tested whether Plasmodium immunotherapy combined with gemcitabine (Gem), a representative chemotherapy drug, has synergistic antitumor effects. Methods We designed subcutaneously and intravenously implanted murine Lewis lung cancer (LLC) models to test the antitumor effect of Plasmodium chabaudi ASS (Pc) infection in combination with Gem treatment and explored its underlying mechanisms. Results We found that both Pc infection alone and Gem treatment alone significantly inhibited tumor growth in the subcutaneous model, and combination therapy was more effective than either monotherapy. Monotherapy only tended to prolong the survival of tumor-bearing mice, while the combination therapy significantly extended the survival of mice, indicating a significant synergistic effect of the combination. In the mechanistic experiments, we found that the combination therapy significantly upregulated E-cadherin and downregulated Snail protein expression levels, thus inhibiting epithelial-mesenchymal transition (EMT) of tumor cells, which may be due to the blockade of CXCR2/TGF-β-mediated PI3K/Akt/GSK-3β signaling pathway. Conclusion The combination of Pc and Gem plays a synergistic role in inhibiting tumor growth and metastasis, and prolonging mice survival in murine lung cancer models. These effects are partially attributed to the inhibition of EMT of tumor cells, which is potentially due to the blockade of CXCR2/TGF-β-mediated PI3K/Akt/GSK-3β/Snail signaling pathway. The clinical transformation of Plasmodium immunotherapy combined with Gem for lung cancer is worthy of expectation.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Medical Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, Guangdong, China
| | - Zhu Tao
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, Guangdong, China
- CAS-Lamvac (Guangzhou) Biomedical Technology Co., Ltd., Guangzhou, Guangdong, China
| | - Yun Liang
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, Guangdong, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Meng Ma
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, Guangdong, China
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Dickson Adah
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, Guangdong, China
| | - Wenting Ding
- CAS-Lamvac (Guangzhou) Biomedical Technology Co., Ltd., Guangzhou, Guangdong, China
| | - Lili Chen
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, Guangdong, China
| | - Xiaofen Li
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, Guangdong, China
| | - Linglin Dai
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, Guangdong, China
| | - Songwe Fanuel
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, Guangdong, China
- Department of Applied Biosciences and Biotechnology, Faculty of Science and Technology, Midlands State University, Gweru, Zimbabwe
| | - Siting Zhao
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, Guangdong, China
- CAS-Lamvac (Guangzhou) Biomedical Technology Co., Ltd., Guangzhou, Guangdong, China
| | - Wen Hu
- CAS-Lamvac (Guangzhou) Biomedical Technology Co., Ltd., Guangzhou, Guangdong, China
| | - Donghai Wu
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, Guangdong, China
| | - Ziyuan Duan
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, Guangdong, China
| | - Fang Zhou
- CAS-Lamvac (Guangzhou) Biomedical Technology Co., Ltd., Guangzhou, Guangdong, China
| | - Li Qin
- CAS-Lamvac (Guangzhou) Biomedical Technology Co., Ltd., Guangzhou, Guangdong, China
| | - Xiaoping Chen
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences (CAS), Guangzhou, Guangdong, China
- CAS-Lamvac (Guangzhou) Biomedical Technology Co., Ltd., Guangzhou, Guangdong, China
| | - Zhaoqing Yang
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
19
|
Wang L, Du C, Jiang B, Chen L, Wang Z. Adjusting the dose of traditional drugs combined with immunotherapy: reshaping the immune microenvironment in lung cancer. Front Immunol 2023; 14:1256740. [PMID: 37901223 PMCID: PMC10600379 DOI: 10.3389/fimmu.2023.1256740] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/05/2023] [Indexed: 10/31/2023] Open
Abstract
Immunotherapy is currently the most promising clinical treatment for lung cancer, not only revolutionizing second-line therapy but now also approved for first-line treatment. However, its clinical efficiency is not high and not all patients benefit from it. Thus, finding the best combination strategy to expand anti-PD-1/PD-L1-based immunotherapy is now a hot research topic. The conventional use of chemotherapeutic drugs and targeted drugs inevitably leads to resistance, toxic side effects and other problems. Recent research, however, suggests that by adjusting the dosage of drugs and blocking the activation of mutational mechanisms that depend on acquired resistance, it is possible to reduce toxic side effects, activate immune cells, and reshape the immune microenvironment of lung cancer. Here, we discuss the effects of different chemotherapeutic drugs and targeted drugs on the immune microenvironment. We explore the effects of adjusting the dosing sequence and timing, and the mechanisms of such responses, and show how the effectiveness and reliability of combined immunotherapy provide improved treatment outcomes.
Collapse
Affiliation(s)
- Linlin Wang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Changqi Du
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Bing Jiang
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Lin Chen
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zibing Wang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| |
Collapse
|
20
|
Sordo-Bahamonde C, Lorenzo-Herrero S, Gonzalez-Rodriguez AP, Martínez-Pérez A, Rodrigo JP, García-Pedrero JM, Gonzalez S. Chemo-Immunotherapy: A New Trend in Cancer Treatment. Cancers (Basel) 2023; 15:cancers15112912. [PMID: 37296876 DOI: 10.3390/cancers15112912] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Chemotherapy has been the basis of advanced cancer treatment for decades. This therapy has largely been considered immunosuppressive, yet accumulated preclinical and clinical evidence shows that certain chemotherapeutic drugs, under defined conditions, may stimulate antitumor immunity and potentiate immune checkpoint inhibitor (ICI)-based therapy. Its effectiveness has been highlighted by recent regulatory approvals of various combinations of chemotherapy with ICIs in several tumors, particularly in some difficult-to-treat cancers. This review discusses the immune modulatory properties of chemotherapy and how they may be harnessed to develop novel chemo-immunotherapy combinations. It also highlights the key determinants of the success of chemo-immunotherapy and provides an overview of the combined chemo-immunotherapies that have been clinically approved.
Collapse
Affiliation(s)
- Christian Sordo-Bahamonde
- Department of Functional Biology, Immunology, Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Seila Lorenzo-Herrero
- Department of Functional Biology, Immunology, Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Ana P Gonzalez-Rodriguez
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Hematology, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
| | - Alejandra Martínez-Pérez
- Department of Functional Biology, Immunology, Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Juan P Rodrigo
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Otolaryngology-Head and Neck Surgery, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Juana M García-Pedrero
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Segundo Gonzalez
- Department of Functional Biology, Immunology, Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
21
|
Koh EK, Lee HR, Son WC, Park GY, Kim J, Bae JH, Park YS. Combinatorial immunotherapy with gemcitabine and ex vivo-expanded NK cells induces anti-tumor effects in pancreatic cancer. Sci Rep 2023; 13:7656. [PMID: 37169953 PMCID: PMC10175562 DOI: 10.1038/s41598-023-34827-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/09/2023] [Indexed: 05/13/2023] Open
Abstract
Pancreatic cancer is difficult to diagnose at the initial stage and is often discovered after metastasis to nearby organs. Gemcitabine is currently used as a standard treatment for pancreatic cancer. However, since chemotherapy for pancreatic cancer has not yet reached satisfactory therapeutic results, adjuvant chemotherapy methods are attempted. It can be expected that combining immune cell therapy with existing anticancer drug combination treatment will prevent cancer recurrence and increase survival rates. We isolated natural killer (NK) cells and co-cultured them with strongly activated autologous peripheral blood mononuclear cells (PBMCs) as feeder cells, activated using CD3 antibody, IFN-r, IL-2, and γ-radiation. NK cells expanded in this method showed greater cytotoxicity than resting NK cells, when co-cultured with pancreatic cancer cell lines. Tumor growth was effectively inhibited in a pancreatic cancer mouse xenograft model. Therapeutic efficacy was increased by using gemcitabine and erlotinib in combination. These findings suggest that NK cells cultured by the method proposed here have excellent anti-tumor activity. We demonstrate that activated NK cells can efficiently inhibit pancreatic tumors when used in combination with gemcitabine-based therapy.
Collapse
Affiliation(s)
- Eun-Kyoung Koh
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, South Korea
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, 50612, South Korea
| | - Hong-Rae Lee
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, South Korea
| | - Woo-Chang Son
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, South Korea
| | - Ga-Young Park
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, South Korea
| | - Juhee Kim
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, South Korea
| | - Jae-Ho Bae
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, 50612, South Korea.
| | - You-Soo Park
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, South Korea.
| |
Collapse
|
22
|
Obradovic A, Ager C, Turunen M, Nirschl T, Khosravi-Maharlooei M, Iuga A, Jackson CM, Yegnasubramanian S, Tomassoni L, Fernandez EC, McCann P, Rogava M, DeMarzo AM, Kochel CM, Allaf M, Bivalacqua T, Lim M, Realubit R, Karan C, Drake CG, Califano A. Systematic elucidation and pharmacological targeting of tumor-infiltrating regulatory T cell master regulators. Cancer Cell 2023; 41:933-949.e11. [PMID: 37116491 PMCID: PMC10193511 DOI: 10.1016/j.ccell.2023.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/13/2022] [Accepted: 04/06/2023] [Indexed: 04/30/2023]
Abstract
Due to their immunosuppressive role, tumor-infiltrating regulatory T cells (TI-Tregs) represent attractive immuno-oncology targets. Analysis of TI vs. peripheral Tregs (P-Tregs) from 36 patients, across four malignancies, identified 17 candidate master regulators (MRs) as mechanistic determinants of TI-Treg transcriptional state. Pooled CRISPR-Cas9 screening in vivo, using a chimeric hematopoietic stem cell transplant model, confirmed the essentiality of eight MRs in TI-Treg recruitment and/or retention without affecting other T cell subtypes, and targeting one of the most significant MRs (Trps1) by CRISPR KO significantly reduced ectopic tumor growth. Analysis of drugs capable of inverting TI-Treg MR activity identified low-dose gemcitabine as the top prediction. Indeed, gemcitabine treatment inhibited tumor growth in immunocompetent but not immunocompromised allografts, increased anti-PD-1 efficacy, and depleted MR-expressing TI-Tregs in vivo. This study provides key insight into Treg signaling, specifically in the context of cancer, and a generalizable strategy to systematically elucidate and target MR proteins in immunosuppressive subpopulations.
Collapse
Affiliation(s)
- Aleksandar Obradovic
- Columbia Center for Translational Immunology, Irving Medical Center, New York, NY, USA; Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Casey Ager
- Columbia Center for Translational Immunology, Irving Medical Center, New York, NY, USA; Department of Hematology Oncology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mikko Turunen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Thomas Nirschl
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Alina Iuga
- Department of Pathology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Christopher M Jackson
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Lorenzo Tomassoni
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ester Calvo Fernandez
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Patrick McCann
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Meri Rogava
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Angelo M DeMarzo
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christina M Kochel
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mohamad Allaf
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Trinity Bivalacqua
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, CA, USA
| | - Ronald Realubit
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA; J.P. Sulzberger Columbia Genome Center, Columbia University, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Charles Karan
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA; J.P. Sulzberger Columbia Genome Center, Columbia University, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Charles G Drake
- Columbia Center for Translational Immunology, Irving Medical Center, New York, NY, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrea Califano
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; J.P. Sulzberger Columbia Genome Center, Columbia University, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA; Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, NY, USA; Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
23
|
Gong J, Shi T, Liu J, Pei Z, Liu J, Ren X, Li F, Qiu F. Dual-drug codelivery nanosystems: An emerging approach for overcoming cancer multidrug resistance. Biomed Pharmacother 2023; 161:114505. [PMID: 36921532 DOI: 10.1016/j.biopha.2023.114505] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Multidrug resistance (MDR) promotes tumor recurrence and metastasis and heavily reduces anticancer efficiency, which has become a primary reason for the failure of clinical chemotherapy. The mechanisms of MDR are so complex that conventional chemotherapy usually fails to achieve an ideal therapeutic effect and even accelerates the occurrence of MDR. In contrast, the combination of chemotherapy with dual-drug has significant advantages in tumor therapy. A novel dual-drug codelivery nanosystem, which combines dual-drug administration with nanotechnology, can overcome the application limitation of free drugs. Both the characteristics of nanoparticles and the synergistic effect of dual drugs contribute to circumventing various drug-resistant mechanisms in tumor cells. Therefore, developing dual-drug codelivery nanosystems with different multidrug-resistant mechanisms has an important reference value for reversing MDR and enhancing the clinical antitumor effect. In this review, the advantages, principles, and common codelivery nanocarriers in the application of dual-drug codelivery systems are summarized. The molecular mechanisms of MDR and the dual-drug codelivery nanosystems designed based on different mechanisms are mainly introduced. Meanwhile, the development prospects and challenges of codelivery nanosystems are also discussed, which provide guidelines to exploit optimized combined chemotherapy strategies in the future.
Collapse
Affiliation(s)
- Jianing Gong
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Taoran Shi
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jinfeng Liu
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zerong Pei
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingbo Liu
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, China
| | - Xiaoliang Ren
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Fengyun Li
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
24
|
Tulyte S, Characiejus D, Matuzeviciene R, Janiulioniene A, Radzevicius M, Jasiunaite E, Zvirblis T, Sileikis A. Prognostic value of circulating T-lymphocyte subsets in advanced pancreatic cancer patients treated with mFOLFIRINOX or gemcitabine. Int Immunopharmacol 2023; 115:109722. [PMID: 37724957 DOI: 10.1016/j.intimp.2023.109722] [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: 11/26/2022] [Revised: 01/08/2023] [Accepted: 01/08/2023] [Indexed: 01/21/2023]
Abstract
Advanced pancreatic ductal adenocarcinoma (PDAC) is commonly treated with a chemotherapy combination of mFOLFIRINOX or gemcitabine. However, predictive and prognostic factors for choosing a more appropriate treatment strategy are still lacking. This study aimed to evaluate how chemotherapy changes immune system parameters and whether these changes influence survival outcomes. We sought to identify an easily accessible marker to help choose the appropriate treatment. Patients with PDAC who were suitable for systemic chemotherapy were eligible for the study. Peripheral blood samples were obtained at baseline and after two months of treatment. Lymphocyte subsets were measured using flow cytometry. Correlation with clinical features and survival analyses were performed. In total, 124 patients were enrolled in this study. Seventy patients were treated with mFOLFIRINOX and 50 with gemcitabine monotherapy. Four patients could not be treated because of rapid deterioration. During overall survival analysis (OS), significant factors included age, Eastern Cooperative Oncology Group (ECOG) performance status, differentiation grade G3, carcinoma antigen (CA) 19-9 more than 100 kU/L, absolute white blood cell count, CD3 + CD8+, and CD8 + CD57-T lymphocytes. Natural killer CD3-CD56 + CD16 + and CD3-CD56 + CD16- and T regulatory CD4 + FOXP3 + and CD3 + CD56 + cells differed during treatment, but these differences did not influence the survival results. At baseline, CD8 + CD57- T lymphocyte count demonstrated a clear independent impact on progression-free survival and OS. Gemcitabine showed better survival in patients with extremely low baseline CD8 + CD57- levels. Therefore, circulating CD3 + CD8 + and CD8 + CD57- cells measured before treatment in PDAC may be considered prognostic and predictive biomarkers.
Collapse
Affiliation(s)
- Skaiste Tulyte
- Clinic of Internal Diseases, Family Medicine and Oncology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania.
| | - Dainius Characiejus
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Reda Matuzeviciene
- Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Ausra Janiulioniene
- Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Mantas Radzevicius
- Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | | | - Tadas Zvirblis
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Audrius Sileikis
- Clinic of Gastroenterology, Nephrourology and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
25
|
Du Z, Yin H, Zhao S, Ma Y, Sun Z, Dong B, Zhu M, Zhu C, Peng J, Yang T. Case report: Gemcitabine intravesical hyperthermic infusion combined with tislelizumab in muscle invasive bladder urothelium carcinoma. Front Oncol 2022; 12:1062655. [PMID: 36620538 PMCID: PMC9816863 DOI: 10.3389/fonc.2022.1062655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/12/2022] [Indexed: 12/25/2022] Open
Abstract
Background Muscle invasive bladder urothelium carcinoma is a common urinary tract tumor. With the deepening of research, more and more treatment methods are applied in clinical practice, extending the life of patients. Among them, the clinical application of chemotherapeutic intravesical hyperthermia and tumor immunotherapy provides new ideas for our treatment. Case report An 81-year-old female patient was diagnosed with stage T2N0M0 bladder cancer in our hospital. Because the patient and her family were keen to preserve her bladder, they declined surgery and opted for combined chemotherapy. After informed consent from the patient and her family, she received cisplatin combined with gemcitabine intravesical hyperthermic infusion. But the side effects of cisplatin made her intolerable to chemotherapy. With their informed consent we changed her to intravenous tislelizumab in combination with gemcitabine intravesical hyperthermic infusion to continue her treatment. During the subsequent follow-up visits, we found a surprising effect of the treatment. Conclusion Gemcitabine intravesical hyperthermia therapy combined with intravenous tislelizumab in the treatment of muscle invasive bladder urothelium carcinoma may provide a new possible therapeutic strategy of some patients who are inoperable or refuse surgery.
Collapse
|
26
|
de la Cruz-Merino L, Gion M, Cruz J, Alonso-Romero JL, Quiroga V, Moreno F, Andrés R, Santisteban M, Ramos M, Holgado E, Cortés J, López-Miranda E, Cortés A, Henao F, Palazón-Carrión N, Rodriguez LM, Ceballos I, Soto A, Puertes A, Casas M, Benito S, Chiesa M, Bezares S, Caballero R, Jiménez-Cortegana C, Sánchez-Margalet V, Rojo F. Pembrolizumab in combination with gemcitabine for patients with HER2-negative advanced breast cancer: GEICAM/2015-04 (PANGEA-Breast) study. BMC Cancer 2022; 22:1258. [PMID: 36463104 PMCID: PMC9719636 DOI: 10.1186/s12885-022-10363-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 11/24/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND We evaluated a new chemoimmunotherapy combination based on the anti-PD1 monoclonal antibody pembrolizumab and the pyrimidine antimetabolite gemcitabine in HER2- advanced breast cancer (ABC) patients previously treated in the advanced setting, in order to explore a potential synergism that could eventually obtain long term benefit in these patients. METHODS HER2-negative ABC patients received 21-day cycles of pembrolizumab 200 mg (day 1) and gemcitabine (days 1 and 8). A run-in-phase (6 + 6 design) was planned with two dose levels (DL) of gemcitabine (1,250 mg/m2 [DL0]; 1,000 mg/m2 [DL1]) to determine the recommended phase II dose (RP2D). The primary objective was objective response rate (ORR). Tumor infiltrating lymphocytes (TILs) density and PD-L1 expression in tumors and myeloid-derived suppressor cells (MDSCs) levels in peripheral blood were analyzed. RESULTS Fourteen patients were treated with DL0, resulting in RP2D. Thirty-six patients were evaluated during the first stage of Simon's design. Recruitment was stopped as statistical assumptions were not met. The median age was 52; 21 (58%) patients had triple-negative disease, 28 (78%) visceral involvement, and 27 (75%) ≥ 2 metastatic locations. Progression disease was observed in 29 patients. ORR was 15% (95% CI, 5-32). Eight patients were treated ≥ 6 months before progression. Fourteen patients reported grade ≥ 3 treatment-related adverse events. Due to the small sample size, we did not find any clear association between immune tumor biomarkers and treatment efficacy that could identify a subgroup with higher probability of response or better survival. However, patients that experienced a clinical benefit showed decreased MDSCs levels in peripheral blood along the treatment. CONCLUSION Pembrolizumab 200 mg and gemcitabine 1,250 mg/m2 were considered as RP2D. The objective of ORR was not met; however, 22% patients were on treatment for ≥ 6 months. ABC patients that could benefit of chemoimmunotherapy strategies must be carefully selected by robust and validated biomarkers. In our heavily pretreated population, TILs, PD-L1 expression and MDSCs levels could not identify a subgroup of patients for whom the combination of gemcitabine and pembrolizumab would induce long term benefit. TRIAL REGISTRATION ClinicalTrials.gov and EudraCT (NCT03025880 and 2016-001,779-54, respectively). Registration dates: 20/01/2017 and 18/11/2016, respectively.
Collapse
Affiliation(s)
- L. de la Cruz-Merino
- grid.411375.50000 0004 1768 164XDepartment of Medical Oncology, Medicine Department, Virgen Macarena University Hospital, University of Seville, Dr. Fedriani St, No. 3, Seville, 41009 Spain ,grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain
| | - M. Gion
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain ,grid.411347.40000 0000 9248 5770Department of Medical Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - J. Cruz
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain ,grid.411220.40000 0000 9826 9219Department of Medical Oncology, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - JL. Alonso-Romero
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain ,grid.411372.20000 0001 0534 3000Department of Medical Oncology, Hospital Clínico Universitario Virgen de La Arrixaca-IMIB, Murcia, Spain
| | - V. Quiroga
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain ,grid.418701.b0000 0001 2097 8389Department of Medical Oncology, Badalona Applied Research Group in Oncology (B-ARGO Group), Catalan Institute of Oncology, Badalona, Spain
| | - F. Moreno
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain ,grid.411068.a0000 0001 0671 5785Department of Medical Oncology, Hospital Clínico Universitario San Carlos, Madrid, Spain
| | - R. Andrés
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain ,grid.411050.10000 0004 1767 4212Department of Medical Oncology, Hospital Clínico Universitario Lozano Blesa, Saragossa, Spain
| | - M. Santisteban
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain ,grid.411730.00000 0001 2191 685XDepartment of Medical Oncology, Clínica Universidad de Navarra, Navarra, Spain ,grid.508840.10000 0004 7662 6114IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - M. Ramos
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain ,grid.418394.3Department of Medical Oncology, Centro Oncológico de Galicia, A Coruña, Spain
| | - E. Holgado
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain ,grid.414808.10000 0004 1772 3571Department of Medical Oncology, Hospital La Luz, Quironsalud, Madrid, Spain
| | - J. Cortés
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain ,International Breast Cancer Center (IBCC), Quiron Group, Barcelona and Madrid, Spain ,grid.411083.f0000 0001 0675 8654Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain ,grid.119375.80000000121738416Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
| | - E. López-Miranda
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain ,grid.411347.40000 0000 9248 5770Department of Medical Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - A. Cortés
- grid.411347.40000 0000 9248 5770Department of Medical Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - F. Henao
- grid.411375.50000 0004 1768 164XDepartment of Medical Oncology, Medicine Department, Virgen Macarena University Hospital, University of Seville, Dr. Fedriani St, No. 3, Seville, 41009 Spain ,grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain
| | - N. Palazón-Carrión
- grid.411375.50000 0004 1768 164XDepartment of Medical Oncology, Medicine Department, Virgen Macarena University Hospital, University of Seville, Dr. Fedriani St, No. 3, Seville, 41009 Spain ,grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain
| | - L. M. Rodriguez
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain ,grid.411220.40000 0000 9826 9219Department of Medical Oncology, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - I. Ceballos
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain ,grid.411220.40000 0000 9826 9219Department of Medical Oncology, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - A. Soto
- grid.411372.20000 0001 0534 3000Department of Medical Oncology, Hospital Clínico Universitario Virgen de La Arrixaca-IMIB, Murcia, Spain
| | - A. Puertes
- grid.411372.20000 0001 0534 3000Department of Medical Oncology, Hospital Clínico Universitario Virgen de La Arrixaca-IMIB, Murcia, Spain
| | - M. Casas
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain
| | - S. Benito
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain
| | - M. Chiesa
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain
| | - S. Bezares
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain
| | - R. Caballero
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain
| | - C. Jiménez-Cortegana
- grid.411375.50000 0004 1768 164XMedical Biochemistry and Molecular Biology and Immunology Department, Virgen Macarena University Hospital, University of Seville, Seville, Spain
| | - V. Sánchez-Margalet
- grid.411375.50000 0004 1768 164XMedical Biochemistry and Molecular Biology and Immunology Department, Virgen Macarena University Hospital, University of Seville, Seville, Spain
| | - F. Rojo
- grid.430580.aGEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, Madrid, Spain ,grid.419651.e0000 0000 9538 1950Pathology Department, IIS-Fundación Jiménez Díaz, Madrid, Spain ,CIBERONC-ISCIII, Madrid, Spain
| |
Collapse
|
27
|
Hagiwara T, Numano T, Hara T, Sugiyama T, Mera Y, Tamano S, Miyata H. Chemical-induced lung tumor in Tg-rasH2 mice: a novel mouse tumor model to assess immune checkpoint inhibitors combined with a chemotherapy drug. J Toxicol Pathol 2022; 35:321-331. [PMID: 36406167 PMCID: PMC9647217 DOI: 10.1293/tox.2022-0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/06/2022] [Indexed: 09/08/2024] Open
Abstract
In subcutaneous tumor models, changes in the tumor microenvironment can lead to differences in therapeutic treatment responses between the subcutaneous and parent tumors. Accordingly, we generated a lung carcinogenesis model that combines genetically modified mice (Tg-rasH2 mice) with two-stage chemical carcinogenesis as an alternative to the subcutaneous tumor model. In this model, Tg-rasH2 mice were treated with 1-ethyl-1-nitrosourea, followed by butylhydroxytoluene. Mice developed lung adenomas five weeks after treatment initiation. Subsequently, anti-mouse PD-1 antibody (α-mPD-1) or isotype control was administered intraperitoneally twice a week for 4 weeks. Tumor growth was examined by measuring the relative tumor area in serially sliced lung histopathological specimens. No statistically significant differences were observed in the relative lung tumor areas between treated and control groups. A second experiment then examined the antitumor efficacy of α-mPD-1 combined with gemcitabine in a mouse model. Mice were treated identically as in Experiment 1, except that the treated group received once-weekly intraperitoneal injections of 10 mg/kg gemcitabine. In contrast to Experiment 1, the combined treatment significantly reduced the relative tumor areas in the lungs. This result also resembles that of a phase III clinical trial (ORIENT-12), showing that patients with non-small-cell lung carcinoma benefited from combination treatment with gemcitabine and the anti-human PD-1 antibody sintilimab. Thus, this mouse model could be a feasible means to preclinically evaluate the antitumor efficacy of different immunotherapy and chemotherapy drug combinations.
Collapse
Affiliation(s)
- Teruaki Hagiwara
- DIMS Institute of Medical Science, Inc., 64 Goura, Nishiazai, Azai-cho, Ichinomiya-shi, Aichi 491-0113, Japan
| | - Takamasa Numano
- DIMS Institute of Medical Science, Inc., 64 Goura, Nishiazai, Azai-cho, Ichinomiya-shi, Aichi 491-0113, Japan
| | - Tomomi Hara
- DIMS Institute of Medical Science, Inc., 64 Goura, Nishiazai, Azai-cho, Ichinomiya-shi, Aichi 491-0113, Japan
| | - Taiki Sugiyama
- DIMS Institute of Medical Science, Inc., 64 Goura, Nishiazai, Azai-cho, Ichinomiya-shi, Aichi 491-0113, Japan
| | - Yukinori Mera
- DIMS Institute of Medical Science, Inc., 64 Goura, Nishiazai, Azai-cho, Ichinomiya-shi, Aichi 491-0113, Japan
| | - Seiko Tamano
- DIMS Institute of Medical Science, Inc., 64 Goura, Nishiazai, Azai-cho, Ichinomiya-shi, Aichi 491-0113, Japan
| | - Hiroto Miyata
- DIMS Institute of Medical Science, Inc., 64 Goura, Nishiazai, Azai-cho, Ichinomiya-shi, Aichi 491-0113, Japan
| |
Collapse
|
28
|
Fu G, Wu Y, Zhao G, Chen X, Xu Z, Sun J, Tian J, Cheng Z, Shi Y, Jin B. Activation of cGAS-STING Signal to Inhibit the Proliferation of Bladder Cancer: The Immune Effect of Cisplatin. Cells 2022; 11:cells11193011. [PMID: 36230972 PMCID: PMC9564335 DOI: 10.3390/cells11193011] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/09/2022] [Accepted: 09/22/2022] [Indexed: 11/29/2022] Open
Abstract
Cisplatin is commonly used in neoadjuvant, adjuvant, and systemic therapy for advanced bladder cancer, but its immune-related mechanism is still unclear. Exploration of the immune effects of cisplatin in bladder cancer would complement the comprehensive mechanism of cisplatin and provide the basis for combination therapy of cisplatin and immunotherapy in bladder cancer. We confirmed the immune effects of cisplatin on T24 and TCCSUP bladder cancer cell lines in vitro and explored the important function of these immune effects in the bladder cancer microenvironment in a mice tumor model. We found cisplatin induced immune response in bladder cancer by RNA sequencing and validated that cGAS-STING signal was deeply involved in this response. Cisplatin induced cGAS-STING signal inhibited the proliferation of bladder cancer and increased the infiltration percentages of CD8+ T cells and dendritic cells in a transplantation mice tumor model. Accumulation of dsDNA and the release of chromatin bound cGAS are important to activate downstream STING. Our findings indicated a cisplatin-related immune effect in bladder cancer, and cisplatin combined with immunotherapy might have a synergistic effect for bladder cancer therapy.
Collapse
|
29
|
Jiao M, Liu H, Liu X. Transcriptional patterns reveal tumor histologic heterogeneity and immunotherapy response in lung adenocarcinoma. Front Immunol 2022; 13:957751. [PMID: 36003401 PMCID: PMC9393366 DOI: 10.3389/fimmu.2022.957751] [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: 05/31/2022] [Accepted: 06/27/2022] [Indexed: 12/02/2022] Open
Abstract
Tumoral heterogeneity has proven to be a leading cause of difference in prognosis and acquired drug resistance. High intratumor heterogeneity often means poor clinical response and prognosis. Histopathological subtypes suggest tumor heterogeneity evolved during the progression of lung adenocarcinoma, but the exploration of its molecular mechanisms remains limited. In this work, we first verified that transcriptional patterns of a set of differentially expressed genes profoundly revealed the histologic progression of lung adenocarcinoma. Next, a predictive model based on the transcriptional patterns was established to accurately distinguish histologic subtypes. Two crucial genes were identified and used to construct a tumor heterogeneous scoring model (L2SITH) to stratify patients, and we found that patients with low heterogeneity score had better prognosis. Low L2SITH scores implied low tumor purity and beneficial tumor microenvironment. Moreover, L2SITH effectively identified cohorts with better responses to anti–PD-1 immunotherapy.
Collapse
Affiliation(s)
| | - Hui Liu
- *Correspondence: Hui Liu, ; Xuejun Liu,
| | | |
Collapse
|
30
|
Principe DR, Kamath SD, Korc M, Munshi HG. The immune modifying effects of chemotherapy and advances in chemo-immunotherapy. Pharmacol Ther 2022; 236:108111. [PMID: 35016920 PMCID: PMC9271143 DOI: 10.1016/j.pharmthera.2022.108111] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/06/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment paradigm for several malignancies. While the use of single-agent or combined ICIs has achieved acceptable disease control rates in a variety of solid tumors, such approaches have yet to show substantial therapeutic efficacy in select difficult-to-treat cancer types. Recently, select chemotherapy regimens are emerging as extensive modifiers of the tumor microenvironment, leading to the reprogramming of local immune responses. Accordingly, data is now emerging to suggest that certain anti-neoplastic agents modulate various immune cell processes, most notably the cross-presentation of tumor antigens, leukocyte trafficking, and cytokine biosynthesis. As such, the combination of ICIs and cytotoxic chemotherapy are beginning to show promise in many cancers that have long been considered poorly responsive to ICI-based immunotherapy. Here, we discuss past and present attempts to advance chemo-immunotherapy in these difficult-to-treat cancer histologies, mechanisms through which select chemotherapies modify tumor immunogenicity, as well as important considerations when designing such approaches to maximize efficacy and improve therapeutic response rates.
Collapse
Affiliation(s)
- Daniel R Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL, USA; Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL, USA.
| | - Suneel D Kamath
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - Murray Korc
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Hidayatullah G Munshi
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
31
|
Huang H, Peng L, Zhang B, Till BG, Yang Y, Zhang X, Zhao L, Fu X, Li T, Han L, Qin P, Chen L, Yan X, Liu Y, Wang W, Ye Z, Li H, Gao Q, Wang Z. Combination of Low-Dose Gemcitabine and PD-1 Inhibitors for Treatment in Patients With Advanced Malignancies. Front Immunol 2022; 13:882172. [PMID: 35911715 PMCID: PMC9328170 DOI: 10.3389/fimmu.2022.882172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose This study determined the efficacy of low-dose gemcitabine combined with programmed death-1 (PD-1) inhibitors for treating multiple malignancies, providing a cost-effective and safe treatment option. Study Design This study included 61 patients with advanced solid tumors treated with low-dose gemcitabine combined with PD-1 inhibitors at the Henan Cancer Hospital between January 2018 and February 2022. We retrospectively reviewed medical records to evaluate several clinical factors, including progression-free survival (PFS), overall survival (OS), adverse effects (AEs), and objective response to treatment. Results Sixty-one patients received treatment with low-dose gemcitabine combined with PD-1 inhibitors. The objective response rate (ORR) was 29.5% and the disease control rate (DCR) was 62.3%. The median PFS was 4.3 months (95% confidence interval, 2.3 to 6.3 months) and the median OS was 15.0 months (95% confidence interval, 8.8 to 21.2 months). Hematological toxicity, mainly leukopenia or thrombocytopenia, was the most common AE, with any-grade and grade 3/4 hematological toxicity reported in 60.7 and 13.1% of patients, respectively. Conclusions Low-dose gemcitabine combined with PD-1 inhibitors may offer a novel treatment option for patients with advanced malignancies.
Collapse
Affiliation(s)
- Hao Huang
- Department of Immunotherapy, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Ling Peng
- Department of Respiratory Disease, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Bicheng Zhang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Brian G. Till
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Yonghao Yang
- Department of Immunotherapy, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xiaojie Zhang
- Department of Immunotherapy, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Lingdi Zhao
- Department of Immunotherapy, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xiaomin Fu
- Department of Immunotherapy, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Tiepeng Li
- Department of Immunotherapy, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Lu Han
- Department of Immunotherapy, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Peng Qin
- Department of Immunotherapy, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Lin Chen
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health GZMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xiang Yan
- Medical Oncology Department, Chinese People's Liberation Army PLA General Hospital, Beijing, China
| | - Yang Liu
- Department of Radiotherapy, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Wenkang Wang
- Department of Breast Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenlong Ye
- Mengchao Cancer Hospital, Shanghai University, Shanghai, China
- Department of Immune Cell Research, Shanghai Engineering Research Center for Cell Therapy, Shanghai, China
- School of Pharmacy, Binzhou Medical University, Binzhou, China
| | - Hongle Li
- Molecular Pathology Department, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- *Correspondence: Zibing Wang, ; Quanli Gao, ; Hongle Li,
| | - Quanli Gao
- Department of Immunotherapy, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- *Correspondence: Zibing Wang, ; Quanli Gao, ; Hongle Li,
| | - Zibing Wang
- Department of Immunotherapy, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- *Correspondence: Zibing Wang, ; Quanli Gao, ; Hongle Li,
| |
Collapse
|
32
|
Joshi S, Sharabi A. Targeting myeloid-derived suppressor cells to enhance natural killer cell-based immunotherapy. Pharmacol Ther 2022; 235:108114. [DOI: 10.1016/j.pharmthera.2022.108114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 12/09/2022]
|
33
|
Franklin M, Connolly E, Hussell T. Recruited and Tissue-Resident Natural Killer Cells in the Lung During Infection and Cancer. Front Immunol 2022; 13:887503. [PMID: 35844626 PMCID: PMC9284027 DOI: 10.3389/fimmu.2022.887503] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/17/2022] [Indexed: 11/23/2022] Open
Abstract
Natural killer (NK) cells are an important component of the innate immune system, and have a key role in host defense against infection and in tumor surveillance. Tumors and viruses employ remarkably similar strategies to avoid recognition and killing by NK cells and so much can be learnt by comparing NK cells in these disparate diseases. The lung is a unique tissue environment and immune cells in this organ, including NK cells, exist in a hypofunctional state to prevent activation against innocuous stimuli. Upon infection, rapid NK cell infiltration into the lung occurs, the amplitude of which is determined by the extent of inflammation and damage. Activated NK cells kill infected cells and produce pro-inflammatory cytokines and chemokines to recruit cells of the adaptive immune system. More recent evidence has shown that NK cells also play an additional role in resolution of inflammation. In lung cancer however, NK cell recruitment is impaired and those that are present have reduced functionality. The majority of lung NK cells are circulatory, however recently a small population of tissue-resident lung NK cells has been described. The specific role of this subset is yet to be determined, but they show similarity to resident memory T cell subsets. Whether resident or recruited, NK cells are important in the control of pulmonary infections, but equally, can drive excessive inflammation if not regulated. In this review we discuss how NK cells are recruited, controlled and retained in the specific environment of the lung in health and disease. Understanding these mechanisms in the context of infection may provide opportunities to promote NK cell recruitment and function in the lung tumor setting.
Collapse
|
34
|
Jones AB, Rocco A, Lamb LS, Friedman GK, Hjelmeland AB. Regulation of NKG2D Stress Ligands and Its Relevance in Cancer Progression. Cancers (Basel) 2022; 14:2339. [PMID: 35565467 PMCID: PMC9105350 DOI: 10.3390/cancers14092339] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 01/27/2023] Open
Abstract
Under cellular distress, multiple facets of normal homeostatic signaling are altered or disrupted. In the context of the immune landscape, external and internal stressors normally promote the expression of natural killer group 2 member D (NKG2D) ligands that allow for the targeted recognition and killing of cells by NKG2D receptor-bearing effector populations. The presence or absence of NKG2D ligands can heavily influence disease progression and impact the accessibility of immunotherapy options. In cancer, tumor cells are known to have distinct regulatory mechanisms for NKG2D ligands that are directly associated with tumor progression and maintenance. Therefore, understanding the regulation of NKG2D ligands in cancer will allow for targeted therapeutic endeavors aimed at exploiting the stress response pathway. In this review, we summarize the current understanding of regulatory mechanisms controlling the induction and repression of NKG2D ligands in cancer. Additionally, we highlight current therapeutic endeavors targeting NKG2D ligand expression and offer our perspective on considerations to further enhance the field of NKG2D ligand biology.
Collapse
Affiliation(s)
- Amber B. Jones
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Abbey Rocco
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.R.); (G.K.F.)
| | | | - Gregory K. Friedman
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.R.); (G.K.F.)
| | - Anita B. Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| |
Collapse
|
35
|
Maiorano BA, De Giorgi U, Ciardiello D, Schinzari G, Cisternino A, Tortora G, Maiello E. Immune-Checkpoint Inhibitors in Advanced Bladder Cancer: Seize the Day. Biomedicines 2022; 10:biomedicines10020411. [PMID: 35203620 PMCID: PMC8962271 DOI: 10.3390/biomedicines10020411] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/05/2022] [Accepted: 02/06/2022] [Indexed: 02/05/2023] Open
Abstract
Background: In advanced bladder cancer (BCa), platinum-based chemotherapy represents the first-choice treatment. In the last ten years, immune checkpoint inhibitors (ICIs) have changed the therapeutic landscape of many solid tumors. Our review aims to summarize the main findings regarding the clinical use of ICIs in advanced BCa. Methods: We searched PubMed, Embase, and Cochrane databases, and conference abstracts from international congresses (ASCO, ESMO, ASCO GU) for clinical trials, focusing on ICIs as monotherapy and combinations in metastatic BCa. Results: 18 studies were identified. ICIs targeting PD1 (nivolumab, pembrolizumab), PD-L1 (avelumab, atezolizumab, durvalumab), and CTLA4 (ipilimumab, tremelimumab) were used. Survival outcomes have been improved by second-line ICIs, whereas first-line results are dismal. Avelumab maintenance in patients obtaining disease control with chemotherapy has achieved the highest survival rates. Conclusions: ICIs improve survival after platinum-based chemotherapy. Avelumab maintenance represents a new practice-changing treatment. The combinations of ICIs and other compounds, such as FGFR-inhibitors, antibody-drug conjugates, and anti-angiogenic drugs, represent promising therapeutic approaches. Biomarkers with predictive roles and sequencing strategies are warranted for best patient selection.
Collapse
Affiliation(s)
- Brigida Anna Maiorano
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (D.C.); (E.M.)
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (G.S.); (G.T.)
- Correspondence:
| | - Ugo De Giorgi
- Department of Oncology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Davide Ciardiello
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (D.C.); (E.M.)
- Medical Oncology Unit, Department of Precision Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy
| | - Giovanni Schinzari
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (G.S.); (G.T.)
- Comprehensive Cancer Center, Medical Oncology Unit, IRCCS Foundation “A. Gemelli” Policlinic, 00168 Rome, Italy
| | - Antonio Cisternino
- Urology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Giampaolo Tortora
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (G.S.); (G.T.)
- Comprehensive Cancer Center, Medical Oncology Unit, IRCCS Foundation “A. Gemelli” Policlinic, 00168 Rome, Italy
| | - Evaristo Maiello
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (D.C.); (E.M.)
| |
Collapse
|
36
|
Gao X, Liu J, Fan D, Li X, Fang Z, Yan K, Fan Y. Berberine enhances gemcitabine‑induced cytotoxicity in bladder cancer by downregulating Rad51 expression through inactivating the PI3K/Akt pathway. Oncol Rep 2021; 47:33. [PMID: 34935059 DOI: 10.3892/or.2021.8244] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 10/19/2021] [Indexed: 11/09/2022] Open
Affiliation(s)
- Xinghua Gao
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jikai Liu
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Daming Fan
- Department of Pathology, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| | - Xiaofeng Li
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhiqing Fang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Keqiang Yan
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yidong Fan
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
37
|
Chen J, Bao Y, Song Y, Zhang C, Qiu F, Sun Y, Xin L, Cao J, Jiang Y, Luo J, Zhang C, Wang G, Li Q, Liu Y, Tong W, Huang P. Hypoxia-alleviated nanoplatform to enhance chemosensitivity and sonodynamic effect in pancreatic cancer. Cancer Lett 2021; 520:100-108. [PMID: 34245853 DOI: 10.1016/j.canlet.2021.07.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 12/21/2022]
Abstract
Pancreatic cancer is a severe disease that threatens human health. The hypoxic tumor microenvironment in pancreatic cancer leads to resistance to conventional therapies and helps to maintain tumor malignancy. First-line drugs present the disadvantage of systemic side effects, and a synergistic method with sonodynamic therapy (SDT) has been established as an emerging approach. In this study, we produced hypoxia-alleviating nanoplatforms (denoted as PZGI NPs) with zeolitic imidazolate frameworks-90 (ZIF-90) nanoparticles nucleating on platinum (Pt) nanoparticles and co-loaded with gemcitabine and IR780. This platform can catalyze peroxide to oxygen with loaded Pt nanoparticles to alleviate tumor hypoxia. Moreover, the loaded drugs could be quickly released in the lysosome microenvironment, which has a low pH value and high ATP level microenvironment in the mitochondria. This strategy could enhance the sensitivity of cancer cells to chemotherapy. Further, under ultrasound exposure, it could transfer the produced oxygen into a highly cytotoxic singlet oxygen for the augmented sonodynamic effect. Therefore, this multifunctional hypoxia-alleviating nanoplatform offers a promising strategy for chemo-sonodynamic therapy against pancreatic cancer.
Collapse
Affiliation(s)
- Jifan Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yuheng Bao
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310007, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yue Song
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Cong Zhang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Fuqiang Qiu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yu Sun
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Lei Xin
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jing Cao
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yifan Jiang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jiali Luo
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Chao Zhang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Guowei Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Qunyin Li
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yajing Liu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Weijun Tong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310007, China.
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
38
|
Immunogenic senescence sensitizes lung cancer to LUNX-targeting therapy. Cancer Immunol Immunother 2021; 71:1403-1417. [PMID: 34674012 PMCID: PMC9123058 DOI: 10.1007/s00262-021-03077-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/30/2021] [Indexed: 11/02/2022]
Abstract
The higher immunogenicity of tumors usually predicts favorable therapeutic responses. Tumor antigens dominate the immunogenic character within tumors. We investigated if there was a targetable tumor antigen during immunogenic chemotherapy within lung cancer. Chemotherapy-induced immunogenic senescence was demonstrated using a multi-marker, three-step workflow, and RNA-sequencing data. The ability of anti-lung-specific X protein (LUNX) antibody to suppress the survival of senescent lung cancer cells was evaluated in vitro and in vivo using real-time cytotoxicity analysis and xenograft mouse models, respectively. The induction of cellular senescence by immunogenic chemotherapy boosted cell-surface shuttling of LUNX and enhanced the immunogenic features of senescent tumor cells, which sensitized lung cancer cells to anti-LUNX antibody-mediated therapy and contributed to tumor suppression. The immunogenic senescence-mediated anti-tumor response was triggered by the direct action of antibody on tumor cells, strengthened by natural-killer cells through an antibody-dependent cell-mediated cytotoxicity response, and ultimately, led to tumor control. Our findings suggest that LUNX is a lung cancer targetable-immunogenic antigen. The proportion of lung cancers responding to LUNX-targeting therapy could be expanded substantially by immunogenic chemotherapy that induces senescence-associated translocation of LUNX to the plasma membrane.
Collapse
|
39
|
Ramos da Silva J, Ramos Moreno AC, Silva Sales N, de Oliveira Silva M, Aps LRMM, Porchia BFMM, Bitencourt Rodrigues K, Cestari Moreno N, Venceslau-Carvalho AA, Menck CFM, de Oliveira Diniz M, de Souza Ferreira LC. A therapeutic DNA vaccine and gemcitabine act synergistically to eradicate HPV-associated tumors in a preclinical model. Oncoimmunology 2021; 10:1949896. [PMID: 34367730 PMCID: PMC8312617 DOI: 10.1080/2162402x.2021.1949896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Although active immunotherapies are effective strategies to induce activation of CD8+ T cells, advanced stage tumors require further improvements for efficient control. Concerning the burden of cancer-related to Human papillomavirus (HPV), particularly the high incidence and mortality of cervical cancer, our group developed an approach based on a DNA vaccine targeting the HPV-16 E7 oncoprotein (pgDE7h). This immunotherapy is capable of inducing an antitumour CD8+ T cell response but show only partial control of tumors in more advanced growth stages. Here, we combined a chemotherapeutic agent (gemcitabine- Gem) with pgDE7h to overcome immunosuppression and improve antitumour responses in a preclinical mouse tumor model. Our results demonstrated that administration of Gem had synergistic antitumor effects when combined with pgDE7h leading to eradication of both early-stages and established tumors. Overall, the antiproliferative effects of Gem observed in vitro and in vivo provided an optimal window for immunotherapy. In addition, the enhanced antitumour responses induced by the combined therapeutic regimen included enhanced frequencies of antigen-presenting cells (APCs), E7-specific IFN-γ-producing CD8+ T cells, and cytotoxic CD8+ T cells and, concomitantly, less pronounced accumulation of immunosuppressive myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs). These findings demonstrated that the combination of Gem and an active immunotherapy strategy show increased effectiveness, leading to a reduced need for multiple drug doses and, therefore, decreased deleterious side effects avoiding resistance and tumor relapses. Altogether, our results provide evidence for a new and feasible chemoimmunotherapeutic strategy that supports future clinical translation.
Collapse
Affiliation(s)
- Jamile Ramos da Silva
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Ana Carolina Ramos Moreno
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Natiely Silva Sales
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Mariângela de Oliveira Silva
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Luana R M M Aps
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Bruna F M M Porchia
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil.,Laboratory of Tumor Immunology, Department of Immunology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Karine Bitencourt Rodrigues
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Natália Cestari Moreno
- DNA Repair Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil.,Mitochondrial Genetics Lab. Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | - Carlos Frederico M Menck
- DNA Repair Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Mariana de Oliveira Diniz
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil.,Division of Infection and Immunity, University College London, 5 University St, Bloomsbury, London, UK
| | - Luís Carlos de Souza Ferreira
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
40
|
Gaggianesi M, Di Franco S, Pantina VD, Porcelli G, D'Accardo C, Verona F, Veschi V, Colarossi L, Faldetta N, Pistone G, Bongiorno MR, Todaro M, Stassi G. Messing Up the Cancer Stem Cell Chemoresistance Mechanisms Supported by Tumor Microenvironment. Front Oncol 2021; 11:702642. [PMID: 34354950 PMCID: PMC8330815 DOI: 10.3389/fonc.2021.702642] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the recent advances in cancer patient management and in the development of targeted therapies, systemic chemotherapy is currently used as a first-line treatment for many cancer types. After an initial partial response, patients become refractory to standard therapy fostering rapid tumor progression. Compelling evidence highlights that the resistance to chemotherapeutic regimens is a peculiarity of a subpopulation of cancer cells within tumor mass, known as cancer stem cells (CSCs). This cellular compartment is endowed with tumor-initiating and metastasis formation capabilities. CSC chemoresistance is sustained by a plethora of grow factors and cytokines released by neighboring tumor microenvironment (TME), which is mainly composed by adipocytes, cancer-associated fibroblasts (CAFs), immune and endothelial cells. TME strengthens CSC refractoriness to standard and targeted therapies by enhancing survival signaling pathways, DNA repair machinery, expression of drug efflux transporters and anti-apoptotic proteins. In the last years many efforts have been made to understand CSC-TME crosstalk and develop therapeutic strategy halting this interplay. Here, we report the combinatorial approaches, which perturb the interaction network between CSCs and the different component of TME.
Collapse
Affiliation(s)
- Miriam Gaggianesi
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Simone Di Franco
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Vincenzo Davide Pantina
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Gaetana Porcelli
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Caterina D'Accardo
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Francesco Verona
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Veronica Veschi
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | | | - Naida Faldetta
- Department of Surgery, Villa Sofia-Cervello Hospital, Palermo, Italy
| | - Giuseppe Pistone
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Maria Rita Bongiorno
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Matilde Todaro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Giorgio Stassi
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| |
Collapse
|
41
|
Cibula D, Rob L, Mallmann P, Knapp P, Klat J, Chovanec J, Minar L, Melichar B, Hein A, Kieszko D, Pluta M, Spacek J, Bartos P, Wimberger P, Madry R, Markowska J, Streb J, Valha P, Hassan HIB, Pecen L, Galluzzi L, Fucikova J, Hrnciarova T, Hraska M, Bartunkova J, Spisek R. Dendritic cell-based immunotherapy (DCVAC/OvCa) combined with second-line chemotherapy in platinum-sensitive ovarian cancer (SOV02): A randomized, open-label, phase 2 trial. Gynecol Oncol 2021; 162:652-660. [PMID: 34294416 DOI: 10.1016/j.ygyno.2021.07.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/01/2021] [Accepted: 07/03/2021] [Indexed: 11/17/2022]
Abstract
OBJECTIVE DCVAC/OvCa is an active cellular immunotherapy designed to stimulate an immune response against ovarian cancer. We explored the safety and efficacy of DCVAC/OvCa plus carboplatin and gemcitabine in platinum-sensitive ovarian cancer. METHODS In this open-label, parallel-group, phase 2 trial (ClinicalTrials.gov number NCT02107950), patients with platinum-sensitive ovarian cancer relapsing after first-line chemotherapy were randomized to DCVAC/OvCa and chemotherapy or chemotherapy alone. DCVAC/OvCa was administered every 3-6 weeks (10 doses). Endpoints included safety, progression-free survival (PFS; primary efficacy endpoint) and overall survival (OS; secondary efficacy endpoint). RESULTS Between November 2013 and May 2015, 71 patients were randomized to chemotherapy in combination with DCVAC/OvCa or to chemotherapy alone. Treatment-emergent adverse events related to DCVAC/OvCa, leukapheresis and chemotherapy occurred in six (16.2%), two (5.4%), and 35 (94.6%) patients in the DCVAC/OvCa group. Chemotherapy-related events occurred in all patients in the chemotherapy group. Seven patients in the DCVAC/OvCa group were excluded from primary efficacy analyses due to failure to receive ≥1 dose of DCVAC/OvCa. PFS was not improved (hazard ratio [HR] 0.73, 95% confidence interval [CI] 0.42-1.28, P = 0.274, data maturity 78.1%). Median OS was significantly prolonged (by 13.4 months) in the DCVAC/OvCa group (HR 0.38, 95% CI 0.20-0.74, P = 0.003; data maturity 56.3%). A signal for enhanced surrogate antigen-specific T-cell activity was seen with DCVAC/OvCa. CONCLUSIONS DCVAC/OvCa combined with chemotherapy had a favorable safety profile in patients with platinum-sensitive ovarian cancer. DCVAC/OvCa did not improve PFS, but the exploratory analyses revealed OS prolongation and enhanced surrogate antigen-specific T-cell activity.
Collapse
Affiliation(s)
- David Cibula
- First Faculty of Medicine, Charles University and General University Hospital in Prague, Apolinarska 18, Prague 12801, Czech Republic.
| | - Lukas Rob
- Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Srobarova 1150, 100 34 Prague 10-Vinohrady, Czech Republic
| | - Peter Mallmann
- University Hospital of Cologne, Kerpener Str. 34 50931 Cologne, Germany
| | - Pawel Knapp
- Medical University of Bialystok, 24a M. Sklodowskiej-Curie Str., 15-276 Bialystok, Poland
| | - Jaroslav Klat
- Department of Gynecology and Obstetrics, University Hospital Ostrava, 17. listopadu 1790, 708 52 Ostrava, Czech Republic
| | - Josef Chovanec
- Masaryk Memorial Cancer Institute, Zluty kopec 7, 653 53 Brno, Czech Republic
| | - Lubos Minar
- Department of Gynecology and Obstetrics, University Hospital Brno and Masaryk University, Jihlavska 20, 625 00 Brno, Czech Republic
| | - Bohuslav Melichar
- Department of Oncology, Palacky University Medical School and University Hospital, I. P. Pavlova 185/6, 779 00 Olomouc, Czech Republic
| | - Alexander Hein
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich Alexander University of Erlangen-Nuremberg, Universitaetsstrasse 21-23, 91054 Erlangen, Germany
| | - Dariusz Kieszko
- Oncological Center of the Lublin Region, ul. dr K. Jaczewskiego, 720-090 Lublin, Poland
| | - Marek Pluta
- Obstetrics and Gynecology Department, 2nd Faculty of Medicine, University Hospital Motol, Charles University, Prague, V Uvalu 84/1, 150 06 Prague 5, Czech Republic
| | - Jiri Spacek
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Pavel Bartos
- Department of Gynecology and Obstetrics, Hospital Novy Jicin, Purkynova 2138/16, 741 01 Novy Jicin, Czech Republic
| | - Pauline Wimberger
- Department of Gynecology and Obstetrics, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Radoslaw Madry
- Department of Oncology, Gynecological-Oncology, Poznan University of Medical Sciences, Collegium Maius, Fredry 10, 61-701 Poznan, Poland
| | - Janina Markowska
- Department of Oncology, Gynecological-Oncology, Poznan University of Medical Sciences, Collegium Maius, Fredry 10, 61-701 Poznan, Poland
| | - Joanna Streb
- Jagiellonian University Hospital, Jakubowskiego 2, 30-688 Krakow, Poland
| | - Petr Valha
- Department of Gynecology and Obstetrics, Hospital Ceske Budejovice, B. Nemcove 585/54, 370 01 Ceske Budejovice, Czech Republic
| | | | - Ladislav Pecen
- SOTIO a.s., Jankovcova 1518/2, 170 00 Prague 7, Czech Republic; Czech Academy of Sciences, Institute of Computer Science, Pod Vodarenskou vezi 271/2, 182 07 Prague 8, Czech Republic
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, 1300 York Avenue, New York, NY 10065, USA; Caryl and Israel Englander Institute for Precision Medicine, 1300 York Avenue, New York, NY 10065, USA; Department of Dermatology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Université de Paris, 15 Rue de l'Ecole de Médecine, 75006 Paris, France
| | - Jitka Fucikova
- SOTIO a.s., Jankovcova 1518/2, 170 00 Prague 7, Czech Republic
| | - Tereza Hrnciarova
- First Faculty of Medicine, Charles University and General University Hospital in Prague, Apolinarska 18, Prague 12801, Czech Republic; SOTIO a.s., Jankovcova 1518/2, 170 00 Prague 7, Czech Republic
| | - Marek Hraska
- SOTIO a.s., Jankovcova 1518/2, 170 00 Prague 7, Czech Republic
| | | | - Radek Spisek
- SOTIO a.s., Jankovcova 1518/2, 170 00 Prague 7, Czech Republic
| |
Collapse
|
42
|
Zhang S, Liu N, Ma M, Huang H, Handley M, Bai X, Shan F. Methionine enkephalin (MENK) suppresses lung cancer by regulating the Bcl-2/Bax/caspase-3 signaling pathway and enhancing natural killer cell-driven tumor immunity. Int Immunopharmacol 2021; 98:107837. [PMID: 34116288 DOI: 10.1016/j.intimp.2021.107837] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/20/2021] [Accepted: 05/29/2021] [Indexed: 12/26/2022]
Abstract
The aim of this study was to investigate how methionine enkephalin (MENK) regulates the biological behavior of lung cancer cells and to further explore its anti-lung cancer mechanisms in vitro and in vivo. The results showed that MENK enhanced the expression of opioid receptor (OGFr) and induced apoptosis of lung cancer cells by activating the Bcl-1/Bax/caspase-3 signaling pathway in vitro and in vivo. However, the regulatory effects of MENK disappeared after blockade of the OGFr. This confirmed that a prerequisite for the anti-tumor action of MENK is binding to OGFr. Additionally, we observed that MENK treatment enhanced the immunogenicity of lung cancer by enhancing the exposure of calreticulin and high mobility group box 1, and increasing the expression of NKG2D ligands. Further studies showed that MENK treatment increased the expression of natural killer (NK) cell-related cytokines such as granzyme B and interferon-γ and NK cell activation. Thus, we concluded that MENK might inhibit the proliferation of lung cancer cells by activating the Bcl-2/Bax/caspase-3 signaling pathway and enhancing immunogenicity and NK cell-driven tumor immunity.
Collapse
Affiliation(s)
- Shuling Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China; Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Ning Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Mingxing Ma
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Hai Huang
- Department of Orthopedic Oncology, the People's Hospital of Liaoning Province, Shenyang 110016, China
| | - Mike Handley
- Cytocom lnc, 3001 Aloma Ave., Winter Park, FL 32792, USA
| | - Xueli Bai
- Department of Gynecology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110004, China.
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang 110122, China.
| |
Collapse
|
43
|
Grivas P, Agarwal N, Pal S, Kalebasty AR, Sridhar SS, Smith J, Devgan G, Sternberg CN, Bellmunt J. Avelumab first-line maintenance in locally advanced or metastatic urothelial carcinoma: Applying clinical trial findings to clinical practice. Cancer Treat Rev 2021; 97:102187. [PMID: 33839438 DOI: 10.1016/j.ctrv.2021.102187] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/21/2022]
Abstract
Although urothelial carcinoma (UC) is considered a chemotherapy-sensitive tumor, progression-free survival and overall survival (OS) are typically short following standard first-line (1L) platinum-containing chemotherapy in patients with locally advanced or metastatic disease. Immune checkpoint inhibitors (ICIs) have antitumor activity in UC and favorable safety profiles compared with chemotherapy; however, trials of 1L ICI monotherapy or chemotherapy + ICI combinations have not yet shown improved OS vs chemotherapy alone. In addition to direct cytotoxicity, chemotherapy has potential immunogenic effects, providing a rationale for assessing ICIs as switch-maintenance therapy. In the JAVELIN Bladder 100 phase 3 trial, avelumab administered as 1L maintenance with best supportive care (BSC) significantly prolonged OS vs BSC alone in patients with locally advanced or metastatic UC that had not progressed with 1L platinum-containing chemotherapy (median OS, 21.4 vs 14.3 months; hazard ratio, 0.69 [95% CI, 0.56-0.86]; P = 0.001). Efficacy benefits were seen across various subgroups, including recipients of 1L cisplatin- or carboplatin-based chemotherapy, patients with PD-L1+ or PD-L1- tumors, and patients with diverse characteristics. Results from JAVELIN Bladder 100 led to the approval of avelumab as 1L maintenance therapy for patients with locally advanced or metastatic UC that has not progressed with platinum-containing chemotherapy. Avelumab 1L maintenance is also included as a standard of care in treatment guidelines for advanced UC with level 1 evidence. This review summarizes the data that supported these developments and discusses practical considerations for administering avelumab maintenance in clinical practice, including patient selection and treatment management.
Collapse
Affiliation(s)
- Petros Grivas
- University of Washington School of Medicine, Fred Hutchinson Cancer Research Center, Seattle Cancer Care Alliance, Seattle, WA, USA.
| | - Neeraj Agarwal
- University of Utah Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Sumanta Pal
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | | | - Jodi Smith
- EMD Serono, Inc., Rockland, MA, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | | | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine, Meyer Cancer Center, New York, New York, USA
| | - Joaquim Bellmunt
- Beth Israel Deaconess Medical Center and IMIM-PSMAR Lab, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
44
|
Stovgaard ES, Asleh K, Riaz N, Leung S, Gao D, Nielsen LB, Lænkholm AV, Balslev E, Jensen MB, Nielsen D, O Nielsen T. The immune microenvironment and relation to outcome in patients with advanced breast cancer treated with docetaxel with or without gemcitabine. Oncoimmunology 2021; 10:1924492. [PMID: 34026336 PMCID: PMC8118411 DOI: 10.1080/2162402x.2021.1924492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Preclinical studies suggest that some effects of conventional chemotherapy, and in particular, gemcitabine, are mediated through enhanced antitumor immune responses. The objective of this study was to use material from a randomized clinical trial to evaluate whether patients with preexisting immune infiltrates responded better to treatment with gemcitabine + docetaxel (GD) compared to docetaxel alone. Formalin fixed, paraffin-embedded breast cancer tissues from SBG0102 phase 3 trial patients randomly assigned to treatment with GD or docetaxel were used. Immunohistochemical staining for CD8, FOXP3, LAG3, PD-1, PD-L1 and CD163 was performed. Tumor infiltrating lymphocytes (TILs) and tumor associated macrophages were evaluated. Prespecified statistical analyses were performed in a formal prospective-retrospective design. Time to progression was primary endpoint and overall survival secondary endpoint. Correlations between biomarker status and endpoints were evaluated using the Kaplan–Meier method and Cox proportional hazards models. Biomarker data was obtained for 237 patients. There was no difference in treatment effect according to biomarker status for the whole cohort. In planned subgroup analysis by PAM50 subtype, in non-luminal (basal-like and HER2E) breast cancers FOXP3 was a significant predictor of treatment effect with GD compared to docetaxel, with a HR of 0.22 (0.09–0.52) for tumors with low FOXP3 compared to HR 0.92 (0.47–1.80) for high FOXP3 TILs (Pinteraction = 0.01). Immune biomarkers were not predictive of added benefit of gemcitabine in a cohort of mixed breast cancer subtypes. However, in non-luminal breast cancers, patients with low FOXP3+ TILs may have significant benefit from added gemcitabine.
Collapse
Affiliation(s)
- Elisabeth S Stovgaard
- Department of Pathology, Herlev and Gentofte, University of Copenhagen, Herlev, Denmark
| | - Karama Asleh
- Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver, BC, Canada
| | - Nazia Riaz
- Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver, BC, Canada.,Centre for Regenerative Medicine and Stem Cell Research, Aga Khan University, Karachi, Pakistan
| | - Samuel Leung
- Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver, BC, Canada
| | - Dongxia Gao
- Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver, BC, Canada
| | - Lise B Nielsen
- Danish Breast Cancer Cooperative Group, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Anne-Vibeke Lænkholm
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Eva Balslev
- Department of Pathology, Herlev and Gentofte, University of Copenhagen, Herlev, Denmark
| | - Maj-Britt Jensen
- Danish Breast Cancer Cooperative Group, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Dorte Nielsen
- Department of Oncology, Herlev and Gentofte, University of Copenhagen, Herlev, Denmark
| | - Torsten O Nielsen
- Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, Vancouver General Hospital, Vancouver, BC, Canada
| |
Collapse
|
45
|
Immunotherapy Combined with Metronomic Dosing: An Effective Approach for the Treatment of NSCLC. Cancers (Basel) 2021; 13:cancers13081901. [PMID: 33920884 PMCID: PMC8071233 DOI: 10.3390/cancers13081901] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary Non-small cell lung cancer (NSCLC) claims almost 80% of the total lung cancer cases, with the late-stage disease having an estimated median survival time of up to five years. Patients with NSCLC benefit from traditional maximum tolerated dose (MTD) chemotherapy alone or combined with immunotherapy. However, efficacious such treatment options lead to side effects and poor patient quality of life. We show that metronomic (MTR) chemotherapy—based on the daily administration of chemotherapeutics in low, nontoxic doses—could potentially supplement MTD treatment options and indirectly prevent tumor growth leading to efficacy and less toxicity. Importantly when MTR chemotherapy is combined with an immunotherapy anti-PD1 agent, the anticipated efficacy is achieved with less toxicity, thus providing new options for the treatment of NSCLC. Abstract Pioneering studies on tumor and immune cell interactions have highlighted immune checkpoint inhibitors (ICIs) as revolutionizing interventions for the management of NSCLC, typically combined with traditional MTD chemotherapies, which usually lead to toxicities and resistance to treatment. Alternatively, MTR chemotherapy is based on the daily low dose administration of chemotherapeutics, preventing tumor growth indirectly by targeting the tumor microenvironment. The effects of MTR administration of an oral prodrug of gemcitabine (OralGem), alone or with anti-PD1, were evaluated. Relevant in vitro and in vivo models were developed to investigate the efficacy of MTR alone or with immunotherapy and the potential toxicities associated with each dosing scheme. MTR OralGem restricted tumor angiogenesis by regulating thrombospondin-1 (TSP-1) and vascular endothelial growth factor A (VEGFA) expression. MTR OralGem enhanced antitumor immunity by increasing T effector responses and cytokine release, concomitant with dampening regulatory T cell populations. Promising pharmacokinetic properties afforded minimized blood and thymus toxicity and favorable bioavailability upon MTR administration compared to MTD. The combination of MTR OralGem with immunotherapy was shown to be highly efficacious and tolerable, illuminating it as a strong candidate therapeutic scheme for the treatment of NSCLC.
Collapse
|
46
|
Peng X, Chen L, Chen L, Wang B, Wang Y, Zhan X. Chimeric antigen receptor-natural killer cells: Novel insight into immunotherapy for solid tumors (Review). Exp Ther Med 2021; 21:340. [PMID: 33732313 PMCID: PMC7903426 DOI: 10.3892/etm.2021.9771] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
The chimeric antigen receptor (CAR) is an artificially modified fusion protein consisting of an extracellular antigen-binding domain, transmembrane domain and intracellular signalling domain. CAR-T therapy has demonstrated remarkable clinical efficacy in hematological malignancies. However, cytokine release syndrome and other side effects have hindered its application in solid tumors. CAR-natural killer (NK) cells have attracted broad attention due to their safety in clinical applications, their mechanism in recognising cancer cells and the abundance of its clinical specimens. Preclinical and clinical trials of human primary NK cells and NK-92 cell lines demonstrated that CAR-NK cells are able to fight haematological malignancies and solid tumors. However, the implication of CAR-NK cell therapy also has certain challenges, including the expansion and activation of primary NK cells in vitro, selection of CAR targets, survival time of CAR-NK cells in vivo, storage and transportation of NK cells, and efficiency of NK cell transduction. This review focuses on the latest progress of CAR-NK cells in the treatment of solid tumors.
Collapse
Affiliation(s)
- Xiaobo Peng
- Department of Oncology, Changhai Hospital affiliated to Naval Military Medical University, Shanghai 200081, P.R. China
| | - Ling Chen
- Department of Oncology, Changhai Hospital affiliated to Naval Military Medical University, Shanghai 200081, P.R. China
| | - Longpei Chen
- Department of Oncology, Changhai Hospital affiliated to Naval Military Medical University, Shanghai 200081, P.R. China
| | - Bin Wang
- Department of Oncology, Changhai Hospital affiliated to Naval Military Medical University, Shanghai 200081, P.R. China
| | - Yiran Wang
- Department of Oncology, Changhai Hospital affiliated to Naval Military Medical University, Shanghai 200081, P.R. China
| | - Xianbao Zhan
- Department of Oncology, Changhai Hospital affiliated to Naval Military Medical University, Shanghai 200081, P.R. China
| |
Collapse
|
47
|
Zhang S, Zhao J, Bai X, Handley M, Shan F. Biological effects of IL-15 on immune cells and its potential for the treatment of cancer. Int Immunopharmacol 2020; 91:107318. [PMID: 33383444 DOI: 10.1016/j.intimp.2020.107318] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/09/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023]
Abstract
Interleukin-15 (IL-15) has recently emerged as a novel immunomodulatory cytokine in cancer immunotherapy. IL-15 has the potential to reject and destroy cancer cells in the tumor microenvironment by expanding and activating natural killer (NK), natural killer T (NKT), and memory (m) CD8+T cells. Due to the feasible outcomes obtained from preclinical studies and phase 1/2 clinical trials, IL-15-based therapy, including chimeric antigen receptor (CAR) T cell or CAR NK cell infusion following in vitro expansion in the presence of IL-15, used in combination with checkpoint inhibitors and other therapy may extend to clinical practice in the future. It is also important to understand the biological characteristics of IL-15 to ensure the maximal benefit of therapeutic strategies. Here, we summarize the current development of IL-15 in the following areas: anti-tumor mechanisms in the tumor microenvironment, advances in IL-15-based therapy itself or in combination with other methods, including biological agents, monoclonal antibodies, and adoptive immunotherapy.
Collapse
Affiliation(s)
- Shuling Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jianzhu Zhao
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xueli Bai
- Department of Gynecology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110004, China
| | - Mike Handley
- Cytocm lnc, 3001 Aloma Ave, Winter Park, FL 32792, USA
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang 110122, China.
| |
Collapse
|
48
|
Abstract
The rapid development of nanobiotechnology has enabled progress in therapeutic cancer vaccines. These vaccines stimulate the host innate immune response by tumor antigens followed by a cascading adaptive response against cancer. However, an improved antitumor immune response is still in high demand because of the unsatisfactory clinical performance of the vaccine in tumor inhibition and regression. To date, a complicated tumor immunosuppressive environment and suboptimal design are the main obstacles for therapeutic cancer vaccines. The optimization of tumor antigens, vaccine delivery pathways, and proper adjuvants for innate immune response initiation, along with reprogramming of the tumor immunosuppressive environment, is essential for therapeutic cancer vaccines in triggering an adequate antitumor immune response. In this review, we aim to review the challenges in and strategies for enhancing the efficacy of therapeutic vaccines. We start with the summary of the available tumor antigens and their properties and then the optimal strategies for vaccine delivery. Subsequently, the vaccine adjuvants focused on the intrinsic adjuvant properties of nanostructures are further discussed. Finally, we summarize the combination strategies with therapeutic cancer vaccines and discuss their positive impact in cancer immunity.
Collapse
Affiliation(s)
- Jie Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 1001190, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Muhetaerjiang Mamuti
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 1001190, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 1001190, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| |
Collapse
|
49
|
Targeting Tumor-Associated Macrophages in Anti-Cancer Therapies: Convincing the Traitors to Do the Right Thing. J Clin Med 2020; 9:jcm9103226. [PMID: 33050070 PMCID: PMC7600332 DOI: 10.3390/jcm9103226] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/02/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
In the last decade, it has been well-established that tumor-infiltrating myeloid cells fuel not only the process of carcinogenesis through cancer-related inflammation mechanisms, but also tumor progression, invasion, and metastasis. In particular, tumor-associated macrophages (TAMs) are the most abundant leucocyte subset in many cancers and play a major role in the creation of a protective niche for tumor cells. Their ability to generate an immune-suppressive environment is crucial to escape the immune system and to allow the tumor to proliferate and metastasize to distant sites. Conventional therapies, including chemotherapy and radiotherapy, are often not able to limit cancer growth due to the presence of pro-tumoral TAMs; these are also responsible for the failure of novel immunotherapies based on immune-checkpoint inhibition. Several novel therapeutic strategies have been implemented to deplete TAMs; however, more recent approaches aim to use TAMs themselves as weapons to fight cancer. Exploiting their functional plasticity, the reprogramming of TAMs aims to convert immunosuppressive and pro-tumoral macrophages into immunostimulatory and anti-tumor cytotoxic effector cells. This shift eventually leads to the reconstitution of a reactive immune landscape able to destroy the tumor. In this review, we summarize the current knowledge on strategies able to reprogram TAMs with single as well as combination therapies.
Collapse
|