51
|
Yu X, Gao R, Li Y, Zeng C. Regulation of PD-1 in T cells for cancer immunotherapy. Eur J Pharmacol 2020; 881:173240. [PMID: 32497624 DOI: 10.1016/j.ejphar.2020.173240] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/20/2022]
Abstract
Study of the molecular mechanisms underlying cancer immune escape is one of the core issues in immuno-oncology research. Cancer cells can evade T cell cytotoxicity by exploiting the upregulation of T cell inhibitory receptors on T cells and their ligands on cancer cells. These upregulated proteins include the inhibitory receptor programmed cell-death protein 1 (PD-1) and its ligand programmed cell death 1 ligand 1 (PD-L1), which can induce T cell exhaustion and reduce T cell activation. Characterizing PD-1 regulation will help to elucidate the molecular mechanisms underlying T cell exhaustion and improve cancer treatment. Recent studies have found that tumor cells regulate PD-1 during gene transcription, post-transcriptional regulation, and post-translational modification and influence the effects of the anticancer immune response by targeting PD-1. In this review,we summarize the mechanisms of PD-1 regulation in T cells.
Collapse
Affiliation(s)
- Xibao Yu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou, 510632, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory Oncology in South China, Guangzhou, 510060, China
| | - Rili Gao
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou, 510632, China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou, 510632, China.
| | - Chengwu Zeng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
52
|
Zhang Z, Liu J, Zhang C, Li F, Li L, Wang D, Chand D, Guan F, Zang X, Zhang Y. Over-Expression and Prognostic Significance of HHLA2, a New Immune Checkpoint Molecule, in Human Clear Cell Renal Cell Carcinoma. Front Cell Dev Biol 2020; 8:280. [PMID: 32509772 PMCID: PMC7248229 DOI: 10.3389/fcell.2020.00280] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/31/2020] [Indexed: 12/21/2022] Open
Abstract
HHLA2, a newly identified B7 family member, regulates T cell functions. However, the expression and prognostic value of HHLA2 in solid tumors is ill defined. This study aimed to reveal the expression landscape of HHLA2 in various solid tumors, and to evaluate its prognostic value in kidney clear cell carcinoma (KIRC). Using The Cancer Genome Atlas (TCGA) database, we investigated the expression pattern of HHLA2 across 22 types of cancer. HHLA2 and CD8 protein expression was determined via immunohistochemistry (IHC). KIRC-specific findings were further analyzed with R software and the prognostic value was validated on tissue microarrays. HHLA2 was widely expressed in cancers at both the mRNA and protein levels. Among all tested tumors, KIRC showed the highest transcript level of HHLA2, and HHLA2 levels were significantly higher in tumor tissues than in matched normal samples, as evidenced by both TCGA and IHC data. HHLA2 was also positively correlated with survival rates in KIRC based on TCGA and clinical data. Receiver operating characteristic curves data showed the prognostic value of HHLA2 for patients with KIRC in TCGA. Moreover, HHLA2 was positively correlated with immune-related genes, while HHLA2 and CD8 expression exhibited a consistent trend in KIRC tumor samples. In conclusion, HHLA2 is highly expressed in KIRC and predicts a favorable survival outcome, highlighting that it may work as a potential target for KIRC therapy.
Collapse
Affiliation(s)
- Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinyan Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chaoqi Zhang
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feng Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lifeng Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dan Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Damini Chand
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,School of Life Sciences, Zhengzhou University, Zhengzhou, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory for Tumor Immunology and Biotherapy of Henan Province, Zhengzhou, China
| |
Collapse
|
53
|
Selvarajan G. Pembrolizumab: The Nut Cracker. Indian J Med Paediatr Oncol 2020. [DOI: 10.4103/ijmpo.ijmpo_37_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
AbstractAnti-programmed cell death-1 (PD-1)/PD ligand-1 immune checkpoint inhibitors (ICIs) are the newest class of drugs approved for various advanced cancers. Pembrolizumab, an anti-PD1 inhibitor, is approved for treating advanced-stage solid malignancies and refractory lymphomas. Recently, it has been approved as tumor agnostic therapy for microsatellite instability-high advanced-stage disease. In all these studies, pembrolizumab has shown dramatic efficacy with lesser Grade3/4 immune-related adverse events. Contemporarily, immunotherapy paved the way for diagnostic assays and immunotherapy-related response assessment criteria definitions. No published Indian experience with ICIs exists other than isolated case reports. This article aims to review on pembrolizumab mechanism, its indications, and safety. The description of other ICIs is beyond the scope of this review.
Collapse
Affiliation(s)
- Gangothri Selvarajan
- Department of Medical Oncology, Cancer Institute (WIA), Chennai, Tamil Nadu, India
| |
Collapse
|
54
|
Ansa-Addo EA, Huang HC, Riesenberg B, Iamsawat S, Borucki D, Nelson MH, Nam JH, Chung D, Paulos CM, Liu B, Yu XZ, Philpott C, Howe PH, Li Z. RNA binding protein PCBP1 is an intracellular immune checkpoint for shaping T cell responses in cancer immunity. SCIENCE ADVANCES 2020; 6:eaaz3865. [PMID: 32523987 PMCID: PMC7259945 DOI: 10.1126/sciadv.aaz3865] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 03/25/2020] [Indexed: 05/11/2023]
Abstract
Distinct lineages of T cells can act in response to various environmental cues to either drive or restrict immune-mediated pathology. Here, we identify the RNA binding protein, poly(C)-binding protein 1 (PCBP1) as an intracellular immune checkpoint that is up-regulated in activated T cells to prevent conversion of effector T (Teff) cells into regulatory T (Treg) cells, by restricting the expression of Teff cell-intrinsic Treg commitment programs. This was critical for stabilizing Teff cell functions and subverting immune-suppressive signals. T cell-specific deletion of Pcbp1 favored Treg cell differentiation, enlisted multiple inhibitory immune checkpoint molecules including PD-1, TIGIT, and VISTA on tumor-infiltrating lymphocytes, and blunted antitumor immunity. Our results demonstrate a critical role for PCBP1 as an intracellular immune checkpoint for maintaining Teff cell functions in cancer immunity.
Collapse
Affiliation(s)
- Ephraim A. Ansa-Addo
- Department of Microbiology and Immunology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
- Pelotonia Institute for Immuno-Oncology and Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center-James, Columbus, OH 43210, USA
| | - Huai-Cheng Huang
- Department of Microbiology and Immunology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
- National Taiwan University College of Medicine, Graduate Institute of Clinical Medicine, No.7 Chung San South Road, Taipei City 10002, Taiwan
| | - Brian Riesenberg
- Department of Microbiology and Immunology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
- Pelotonia Institute for Immuno-Oncology and Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center-James, Columbus, OH 43210, USA
| | - Supinya Iamsawat
- Department of Microbiology and Immunology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Davis Borucki
- Department of Microbiology and Immunology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michelle H. Nelson
- Department of Microbiology and Immunology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jin Hyun Nam
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Dongjun Chung
- Pelotonia Institute for Immuno-Oncology and Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center-James, Columbus, OH 43210, USA
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Chrystal M. Paulos
- Department of Microbiology and Immunology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Bei Liu
- Department of Microbiology and Immunology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Caroline Philpott
- Genetics and Metabolism Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Bethesda, MD 20892, USA
| | - Philip H. Howe
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Zihai Li
- Department of Microbiology and Immunology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
- Pelotonia Institute for Immuno-Oncology and Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center-James, Columbus, OH 43210, USA
| |
Collapse
|
55
|
Expression of the immune checkpoint VISTA in breast cancer. Cancer Immunol Immunother 2020; 69:1437-1446. [PMID: 32266446 DOI: 10.1007/s00262-020-02554-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 03/23/2020] [Indexed: 12/13/2022]
Abstract
V-domain Ig suppressor of T cell activation (VISTA) is a novel immune checkpoint that is an emerging target for cancer immunotherapy. This study aimed to investigate the expression of VISTA and its association with clinicopathologic parameters as well as with the key immune markers including programmed cell death-1 (PD-1) and PD-1 ligand-1 (PD-L1) in invasive ductal carcinoma (IDC) of the breast [corrected]. Immunohistochemistry was used to detect VISTA, PD-1, PD-L1, and CD8 in tissue microarrays from 919 patients with IDC (N = 341 in the exploratory cohort and = 578 in the validation cohort). VISTA was expressed on the immune cells of 29.1% (267/919) of the samples and on the tumor cells of 8.2% (75/919). VISTA was more frequently expressed in samples that were estrogen receptor-negative, progesterone receptor-negative, human epidermal growth factor receptor 2-positive, poorly differentiated, human epidermal growth factor receptor 2-enriched, and consisting of basal-like tumors. VISTA on immune cells correlated with PD-1, PD-L1, stromal CD8, and tumor-infiltrating lymphocyte expression and was an independent prognostic factor for improved relapse-free and disease-specific survival in patients with estrogen receptor-negative, progesterone receptor-negative, and basal-like IDC. These findings support therapeutic strategies that modulate VISTA expression, perhaps in combination with PD-1/PD-L1 blockade, in human breast cancer immunotherapy.
Collapse
|
56
|
Pan C, Liu H, Robins E, Song W, Liu D, Li Z, Zheng L. Next-generation immuno-oncology agents: current momentum shifts in cancer immunotherapy. J Hematol Oncol 2020; 13:29. [PMID: 32245497 PMCID: PMC7119170 DOI: 10.1186/s13045-020-00862-w] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/23/2020] [Indexed: 12/30/2022] Open
Abstract
Cancer immunotherapy has reached a critical point, now that immune checkpoint inhibitors and two CAR-T products have received market approval in treating 16 types of cancers and 1 tissue-agnostic cancer indication. Accompanying these advances, the 2018 Nobel Prize was awarded for the discovery of immune checkpoint pathways, which has led to the revolution of anti-cancer treatments. However, expanding the indications of immuno-oncology agents and overcoming treatment resistance face mounting challenges. Although combination immunotherapy is an obvious strategy to pursue, the fact that there have been more failures than successes in this effort has served as a wake-up call, placing emphasis on the importance of building a solid scientific foundation for the development of next-generation immuno-oncology (IO) agents. The 2019 China Cancer Immunotherapy Workshop was held to discuss the current challenges and opportunities in IO. At this conference, emerging concepts and strategies for IO development were proposed, focusing squarely on correcting the immunological defects in the tumor microenvironment. New targets such as Siglec-15 and new directions including neoantigens, cancer vaccines, oncolytic viruses, and cytokines were reviewed. Emerging immunotherapies were discussed in the areas of overcoming primary and secondary resistance to existing immune checkpoint inhibitors, activating effector cells, and targeting immunosuppressive mechanisms in the tumor microenvironment. In this article, we highlight old and new waves of IO therapy development, and provide perspectives on the latest momentum shifts in cancer immunotherapy.
Collapse
Affiliation(s)
- Chongxian Pan
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- University of California, Davis, CA, USA
| | - Hongtao Liu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- University of Chicago, Chicago, IL, USA
| | - Elizabeth Robins
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, USA
| | - Wenru Song
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- Kira Pharmaceuticals, Cambridge, MA, USA
| | - Delong Liu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- New York Medical College, Valhalla, NY, USA
| | - Zihai Li
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, USA
| | - Lei Zheng
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.
- Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
57
|
Liu H, Yang Y, Liu Y, Pan J, Wang J, Man F, Zhang W, Liu G. Melanin-Like Nanomaterials for Advanced Biomedical Applications: A Versatile Platform with Extraordinary Promise. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903129. [PMID: 32274309 PMCID: PMC7141020 DOI: 10.1002/advs.201903129] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/31/2019] [Indexed: 05/03/2023]
Abstract
Developing efficient, sustainable, and biocompatible high-tech nanoplatforms derived from naturally existing components in living organisms is highly beneficial for diverse advanced biomedical applications. Melanins are nontoxic natural biopolymers owning widespread distribution in various biosystems, possessing fascinating physicochemical properties and playing significant physiological roles. The multifunctionality together with intrinsic biocompatibility renders bioinspired melanin-like nanomaterials considerably promising as a versatile and powerful nanoplatform with broad bioapplication prospects. This panoramic Review starts with an overview of the fundamental physicochemical properties, preparation methods, and polymerization mechanisms of melanins. A systematical and well-bedded description of recent advancements of melanin-like nanomaterials regarding diverse biomedical applications is then given, mainly focusing on biological imaging, photothermal therapy, drug delivery for tumor treatment, and other emerging biomedicine-related implementations. Finally, current challenges toward clinical translation with an emphasis on innovative design strategies and future striving directions are rationally discussed. This comprehensive and detailed Review provides a deep understanding of the current research status of melanin-like nanomaterials and is expected to motivate further optimization of the design of novel tailorable and marketable multifunctional nanoplatforms in biomedicine.
Collapse
Affiliation(s)
- Heng Liu
- Department of RadiologyPLA Rocket Force Characteristic Medical CenterBeijing100088China
- Department of RadiologyDaping HospitalArmy Medical UniversityChongqing400042China
| | - Youyuan Yang
- Department of RadiologyDaping HospitalArmy Medical UniversityChongqing400042China
| | - Yu Liu
- Department of UltrasoundThe First Affiliated HospitalArmy Medical UniversityChongqing400038China
| | - Jingjing Pan
- Department of RadiologyPLA Rocket Force Characteristic Medical CenterBeijing100088China
| | - Junqing Wang
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityGuangzhou510275China
| | - Fengyuan Man
- Department of RadiologyPLA Rocket Force Characteristic Medical CenterBeijing100088China
| | - Weiguo Zhang
- Department of RadiologyDaping HospitalArmy Medical UniversityChongqing400042China
- Chongqing Clinical Research Center for Imaging and Nuclear MedicineChongqing400042China
| | - Gang Liu
- Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102China
| |
Collapse
|
58
|
Liu M, Gao Y, Yuan Y, Shi S, Yang K, Wu J, Zhang J, Tian J. Global hotspots and future prospects of chimeric antigen receptor T-cell therapy in cancer research: a bibliometric analysis. Future Oncol 2020; 16:597-612. [DOI: 10.2217/fon-2019-0810] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aim: Our study aimed to analyze the characteristics of papers published on CAR T-cell in the field of cancer and explore the existing hot topics and prospects. Materials & methods: We explored the global hotspots and future prospects regarding CAR T-cell therapy in cancer research. Papers of CAR T-cell research were retrieved from the Web of Science database. Analysis was performed using VOSviewer, CiteSpace and Excel software. Results: A total number of 1994 papers related to CAR T-cell research of cancer were included. Molecular Therapy published the most papers (n = 85, 4.26%). A total of 9792 authors participated in the publication of all papers. 62 countries and 2065 institutions have participated in the publication of all papers. Conclusion: Research trends are to improve the immunosuppressive microenvironment of cancer, optimize the structure of CAR T-cells and develop ‘super CAR T-cell’ by using gene-editing technology.
Collapse
Affiliation(s)
- Ming Liu
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, PR China
- Key Laboratory of Evidence-Based Medicine & Knowledge Translation of Gansu Province, Lanzhou 730000, PR China
| | - Ya Gao
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, PR China
- Key Laboratory of Evidence-Based Medicine & Knowledge Translation of Gansu Province, Lanzhou 730000, PR China
| | - Yuan Yuan
- Gansu University of Chinese Medicine, Lanzhou 730000, PR China
| | - Shuzhen Shi
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, PR China
- Key Laboratory of Evidence-Based Medicine & Knowledge Translation of Gansu Province, Lanzhou 730000, PR China
| | - Kelu Yang
- School of Nursing, Lanzhou University, Lanzhou 730000, PR China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100000, PR China
| | - Junhua Zhang
- Evidence-Based Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China
| | - Jinhui Tian
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, PR China
- Key Laboratory of Evidence-Based Medicine & Knowledge Translation of Gansu Province, Lanzhou 730000, PR China
| |
Collapse
|
59
|
Ma M, Lin B, Wang M, Liang X, Su L, Okose O, Lv W, Li J. Immunotherapy in anaplastic thyroid cancer. Am J Transl Res 2020; 12:974-988. [PMID: 32269728 PMCID: PMC7137046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 02/13/2020] [Indexed: 06/11/2023]
Abstract
Anaplastic thyroid cancer (ATC) is one of the worst human malignancies, with an associated median survival of only 5 months. It is resistant to conventional thyroid cancer therapies, including radioiodine and thyroid-stimulating hormone suppression. Cancer immunotherapy has emerged over the past few decades as a transformative approach to treating a wide variety of cancers. However, immunotherapy for ATC is still in the experimental stage. This review will cover several strategies of immunotherapy and discuss the possible application of these strategies in the treatment of ATC (such as targeted therapy for tumor-associated macrophages, cancer vaccines, adoptive immunotherapy, monoclonal antibodies and immune checkpoint blockade) with the hope of improving the prognosis of ATC in the future.
Collapse
Affiliation(s)
- Maoguang Ma
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Bo Lin
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Mingdian Wang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer CenterGuangzhou, China
| | - Xiaoli Liang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Lei Su
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Okenwa Okose
- Texas A & M College of MedicineCollege Station, TX 77843, USA
- Division of Thyroid and Parathyroid Surgery, Massachusetts Eye and Ear Infirmary, Harvard Medical SchoolBoston, MA, USA
| | - Weiming Lv
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Jie Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
- Division of Thyroid and Parathyroid Surgery, Massachusetts Eye and Ear Infirmary, Harvard Medical SchoolBoston, MA, USA
| |
Collapse
|
60
|
Guven DC, Aktas BY, Simsek C, Aksoy S. Gut microbiota and cancer immunotherapy: prognostic and therapeutic implications. Future Oncol 2020; 16:497-506. [PMID: 32100550 DOI: 10.2217/fon-2019-0783] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The immune checkpoint inhibitors have opened new horizons in oncology. Although the indications for the use of Immune checkpoint inhibitors in cancer patients are expanding, there is still a need for markers that can aid in patient selection. Gastrointestinal microbiota can be among these markers. Recently, gastrointestinal microbiota stated to have a bidirectional relation with cancer immunotherapy with roles in both prognostic and therapeutic sides. Preclinical data suggest that modulation of the microbiota could become a novel strategy for improving the efficacy of immunotherapy. However, its labile structure prone to be affected by many factors. Further research can delineate the mechanisms of the relationship between microbiota and immunotherapy can have clinical implications.
Collapse
Affiliation(s)
- Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara 06100, Turkey
| | - Burak Yasin Aktas
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara 06100, Turkey
| | - Cem Simsek
- Department of Gastroenterology, Hacettepe University Faculty of Medicine, Ankara 06100, Turkey
| | - Sercan Aksoy
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara 06100, Turkey
| |
Collapse
|
61
|
Wu D, Lv J, Zhao R, Wu Z, Zheng D, Shi J, Lin S, Wang S, Wu Q, Long Y, Li P, Yao Y. PSCA is a target of chimeric antigen receptor T cells in gastric cancer. Biomark Res 2020; 8:3. [PMID: 32010446 PMCID: PMC6988264 DOI: 10.1186/s40364-020-0183-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/02/2020] [Indexed: 12/23/2022] Open
Abstract
Background Gastric cancer is a deadly malignancy and is a prognostically unfavorable entity with restricted therapeutic strategies available. Prostate stem cell antigen (PSCA) is a glycosylphosphatidylinositol (GPI)-anchored cell surface protein widely expressed in bladder, prostate, and pancreatic cancers. Existing studies have thoroughly recognized the availability of utilizing anti-PSCA CAR-T cells in the treatment of metastatic prostate cancer and non-small-cell lung cancer. However, no previous study has investigated the feasibility of using anti-PSCA CAR-T cells to treat gastric cancer, irrespective of the proven expression of PSCA on the gastric cancer cell surface. Methods We determined the expression of PSCA in several primary tumor tissues and constructed third-generation anti-PSCA CAR-T cells. We then incubated anti-PSCA CAR-T cells and GFP-T cells with target tumor cell lines at E:T ratios of 2:1, 1:1, 1:2, and 1:4 to evaluate the therapeutic efficacy of anti-PSCA CAR-T cells in vitro. We also assayed canonical T cell activation markers after coculturing anti-PSCA CAR-T cells with target cell lines by flow cytometry. The detection of a functional cytokine profile was carried out via enzyme-linked immunosorbent assays. We then evaluated the antitumor activity of anti-PSCA CAR-T cells in vivo by establishing two different xenograft GC mouse models. Results Anti-PSCA CAR-T cells exhibited upregulated activation markers and increased cytokine production profiles related to T cell cytotoxicity in an antigen-dependent manner. Moreover, anti-PSCA CAR-T cells exhibited robust anti-tumor cytotoxicity in vitro. Importantly, we demonstrated that anti-PSCA CAR-T cells delivered by peritumoral injection successfully stunted tumor progression in vivo. However, intravenous administration of anti-PSCA CAR-T cells failed to reveal any therapeutic improvements. Conclusions Our findings corroborated the feasibility of anti-PSCA CAR-T cells and their efficacy against gastric cancer, implicating the potential of applying anti-PSCA CAR-T cells to treat GC patients in the clinic.
Collapse
Affiliation(s)
- Di Wu
- 1School of Life Sciences, University of Science and Technology of China, Hefei, 230027 China.,2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Jiang Lv
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,4University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049 China
| | - Ruocong Zhao
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,5Institute of Hematology, Medical College, Jinan University, Guangzhou, 510632 China
| | - Zhiping Wu
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,4University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049 China
| | - Diwei Zheng
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,4University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049 China
| | - Jingxuan Shi
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,4University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049 China
| | - Simiao Lin
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Suna Wang
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Qiting Wu
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Youguo Long
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Peng Li
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,6Hefei Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Yao Yao
- 2Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,3Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| |
Collapse
|
62
|
Immune biological rationales for the design of combined radio- and immunotherapies. Cancer Immunol Immunother 2020; 69:293-306. [PMID: 31953578 PMCID: PMC7000501 DOI: 10.1007/s00262-019-02460-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 12/22/2019] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapies are promising treatments for many forms of cancer. Nevertheless, the response rates to, e.g., immune checkpoint inhibitors (ICI), are still in low double-digit percentage. This calls for further therapy optimization that should take into account combination of immunotherapies with classical tumor therapies such as radiotherapy. By designing multimodal approaches, immune modulatory properties of certain radiation schemes, additional immune modulation by immunotherapy with ICI and hyperthermia, as well as patient stratification based on genetic and immune constitutions have to be considered. In this context, both the tumor and its microenvironment including cells of the innate and adaptive immune system have to be viewed in synopsis. Knowledge of immune activation and immune suppression by radiation is the basis for well-elaborated addition of certain immunotherapies. In this review, the focus is set on additional immune stimulation by hyperthermia and restoration of an immune response by ICI. The impact of radiation dose and fractionation on immune modulation in multimodal settings has to be considered, as the dynamics of the immune response and the timing between radiotherapy and immunotherapy. Another big challenge is the patient stratification that should be based on matrices of biomarkers, taking into account genetics, proteomics, radiomics, and “immunomics”. One key aim is to turn immunological “cold” tumors into “hot” tumors, and to eliminate barriers of immune-suppressed or immune-excluded tumors. Comprehensive knowledge of immune alterations induced by radiation and immunotherapy when being applied together should be utilized for patient-adapted treatment planning and testing of innovative tumor therapies within clinical trials.
Collapse
|
63
|
Gao S, Wang S, Song Y. Novel immunomodulatory drugs and neo-substrates. Biomark Res 2020; 8:2. [PMID: 31938543 PMCID: PMC6953231 DOI: 10.1186/s40364-020-0182-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/02/2020] [Indexed: 12/12/2022] Open
Abstract
Thalidomide, lenalidomide and pomalidomide are immunomodulatory drugs (IMiDs) effective in the treatment of multiple myeloma, myelodysplastic syndrome (MDS) with deletion of chromosome 5q and other hematological malignancies. Recent studies showed that IMiDs bind to CRBN, a substrate receptor of CRL4 E3 ligase, to induce the ubiquitination and degradation of IKZF1 and IKZF3 in multiple myeloma cells, contributing to their anti-myeloma activity. Similarly, lenalidomide exerts therapeutic efficacy via inducing ubiquitination and degradation of CK1α in MDS with deletion of chromosome 5q. Recently, novel thalidomide analogs have been designed for better clinical efficacy, including CC-122, CC-220 and CC-885. Moreover, a number of neo-substrates of IMiDs have been discovered. Proteolysis-targeting chimeras (PROTACs) as a class of bi-functional molecules are increasingly used as a strategy to target otherwise intractable cellular protein. PROTACs appear to have broad implications for novel therapeutics. In this review, we summarized new generation of immunomodulatory compounds, their potential neo-substrates, and new strategies for the design of novel PROTAC drugs.
Collapse
Affiliation(s)
- Shaobing Gao
- 1The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| | - Shichao Wang
- 2The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfu Front Street, Zhengzhou, 450052 China
| | - Yongping Song
- 1The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| |
Collapse
|
64
|
Di Paolo A, Arrigoni E, Luci G, Cucchiara F, Danesi R, Galimberti S. Precision Medicine in Lymphoma by Innovative Instrumental Platforms. Front Oncol 2019; 9:1417. [PMID: 31921674 PMCID: PMC6928138 DOI: 10.3389/fonc.2019.01417] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 11/28/2019] [Indexed: 12/13/2022] Open
Abstract
In recent years, many efforts have been addressed to the growing field of precision medicine in order to offer individual treatments to every patient on the basis of his/her genetic background. Formerly adopted to achieve new disease classifications as it is still done, innovative platforms, such as microarrays, genome-wide association studies (GWAS), and next generation sequencing (NGS), have made the progress in pharmacogenetics faster and cheaper than previously expected. Several studies in lymphoma patients have demonstrated that these platforms can be used to identify biomarkers predictive of drug efficacy and tolerability, discovering new possible druggable proteins. Indeed, GWAS and NGS allow the investigation of the human genome, finding interesting associations with putative or unexpected targets, which in turns may represent new therapeutic possibilities. Importantly, some objective difficulties have initially hampered the translation of findings in clinical routines, such as the poor quantity/quality of genetic material or the paucity of targets that could be investigated at the same time. At present, some of these technical issues have been partially solved. Furthermore, these analyses are growing in parallel with the development of bioinformatics and its capabilities to manage and analyze big data. Because of pharmacogenetic markers may become important during drug development, regulatory authorities (i.e., EMA, FDA) are preparing ad hoc guidelines and recommendations to include the evaluation of genetic markers in clinical trials. Concerns and difficulties for the adoption of genetic testing in routine are still present, as well as affordability, reliability and the poor confidence of some patients for these tests. However, genetic testing based on predictive markers may offers many advantages to caregivers and patients and their introduction in clinical routine is justified.
Collapse
Affiliation(s)
- Antonello Di Paolo
- Section of Pharmacology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Unit of Clinical Pharmacology and Pharmacogenetics, Pisa University Hospital, Pisa, Italy
| | - Elena Arrigoni
- Section of Pharmacology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giacomo Luci
- Section of Pharmacology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Federico Cucchiara
- Section of Pharmacology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Romano Danesi
- Section of Pharmacology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Unit of Clinical Pharmacology and Pharmacogenetics, Pisa University Hospital, Pisa, Italy
| | - Sara Galimberti
- Section of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Unit of Hematology, Pisa University Hospital, Pisa, Italy
| |
Collapse
|
65
|
Zong L, Zhou Y, Zhang M, Chen J, Xiang Y. VISTA expression is associated with a favorable prognosis in patients with high-grade serous ovarian cancer. Cancer Immunol Immunother 2019; 69:33-42. [PMID: 31781843 PMCID: PMC6949319 DOI: 10.1007/s00262-019-02434-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 11/19/2019] [Indexed: 12/24/2022]
Abstract
Blockading programmed death ligand 1 (PD-L1) shows promising results in patients with some cancers, but not in those with ovarian cancer. V-domain Ig suppressor of T cell activation (VISTA) is a recently discovered immune checkpoint protein that suppresses T cell activation. This study aimed to investigate the expression and clinical significance of VISTA in ovarian cancer as well as its relationship with PD-L1. VISTA and PD-L1 levels in 146 ovarian cancer samples were assessed using immunohistochemistry. We investigated the association between VISTA and other clinicopathological variables, including survival. The associations between the VISTA-encoding C10orf54 gene, other immune checkpoints, and survival were analyzed. VISTA was detected in 51.4% of all samples and 46.6% of PD-L1-negative samples; it was expressed in 28.8%, 35.6%, and 4.1% of tumor cells (TCs), immune cells (ICs), and endothelial cells, respectively. Furthermore, VISTA expression was associated with pathologic type and PD-L1 expression. Moreover, VISTA expression in TCs, but not in ICs, was associated with prolonged progression-free and overall survival in patients with high-grade serous ovarian cancer. The expression of C10orf54 mRNA was associated with prolonged overall survival and immune escape-modulating genes. These results showed that VISTA expression in ovarian tumor cells was associated with a favorable prognosis in patients with high-grade serous ovarian cancer; however, additional studies are required to better understand the expression and role of VISTA in ovarian cancer.
Collapse
Affiliation(s)
- Liju Zong
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.,Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuncan Zhou
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Ming Zhang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Xiang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
66
|
Zhang Z, Yang S, Wang Q. Impact of MET alterations on targeted therapy with EGFR-tyrosine kinase inhibitors for EGFR-mutant lung cancer. Biomark Res 2019; 7:27. [PMID: 31832192 PMCID: PMC6873421 DOI: 10.1186/s40364-019-0179-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/08/2019] [Indexed: 12/24/2022] Open
Abstract
EGFR-tyrosine kinase inhibitors (EGFR-TKIs) have achieved remarkable outcomes in the treatment of patients with EGFR-mutant non-small-cell lung cancer, but acquired resistance is still the main factor restricting their long-term use. In addition to the T790 M mutation of EGFR, amplification of the MET (or c-MET) gene has long been recognized as an important resistance mechanism for first- or second-generation EGFR-TKIs. Recent studies suggest that a key mechanism of acquired resistance to third-generation EGFR-TKIs (such as osimertinib) may be MET amplification and/or protein overactivation, especially when they are used as a first-line treatment. Therefore, in patients resistant to first-generation EGFR-TKIs caused by MET amplification and/or protein overactivation, the combination of osimertinib with MET or MEK inhibitors may be considered.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008 China
| | - Sen Yang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008 China
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008 China
| |
Collapse
|
67
|
Liu D. CAR-T "the living drugs", immune checkpoint inhibitors, and precision medicine: a new era of cancer therapy. J Hematol Oncol 2019; 12:113. [PMID: 31703740 PMCID: PMC6842223 DOI: 10.1186/s13045-019-0819-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
New advances in the design and manufacture of monoclonal antibodies, bispecific T cell engagers, and antibody-drug conjugates make the antibody-directed agents more powerful with less toxicities. Small molecule inhibitors are routinely used now as oral targeted agents for multiple cancers. The discoveries of PD1 and PD-L1 as negative immune checkpoints for T cells have led to the revolution of modern cancer immunotherapy. Multiple agents targeting PD1, PD-L1, or CTLA-4 are widely applied as immune checkpoint inhibitors (ICIs) which alleviate the suppression of immune regulatory machineries and lead to immunoablation of once highly refractory cancers such as stage IV lung cancer. Tisagenlecleucel and axicabtagene ciloleucel are the two approved CD19-targeted chimeric antigen receptor (CAR) T cell products. Several CAR-T cell platforms targeting B cell maturation antigen (BCMA) are under active clinical trials for refractory and/or relapsed multiple myeloma. Still more targets such as CLL-1, EGFR, NKG2D and mesothelin are being directed in CAR-T cell trials for leukemia and solid tumors. Increasing numbers of novel agents are being studied to target cancer-intrinsic oncogenic pathways as well as immune checkpoints. One such an example is targeting CD47 on macrophages which represents a "do-not-eat-me" immune checkpoint. Fueling the current excitement of cancer medicine includes also TCR- T cells, TCR-like antibodies, cancer vaccines and oncolytic viruses.
Collapse
Affiliation(s)
- Delong Liu
- New York Medical College, Valhalla, NY, 10595, USA.
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
68
|
Qin S, Xu L, Yi M, Yu S, Wu K, Luo S. Novel immune checkpoint targets: moving beyond PD-1 and CTLA-4. Mol Cancer 2019; 18:155. [PMID: 31690319 PMCID: PMC6833286 DOI: 10.1186/s12943-019-1091-2] [Citation(s) in RCA: 735] [Impact Index Per Article: 147.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 10/18/2019] [Indexed: 02/10/2023] Open
Abstract
The emergence of immune checkpoint inhibitors (ICIs), mainly including anti-programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) and anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) monoclonal antibodies (mAbs), has shaped therapeutic landscape of some type of cancers. Despite some ICIs have manifested compelling clinical effectiveness in certain tumor types, the majority of patients still showed de novo or adaptive resistance. At present, the overall efficiency of immune checkpoint therapy remains unsatisfactory. Exploring additional immune checkpoint molecules is a hot research topic. Recent studies have identified several new immune checkpoint targets, like lymphocyte activation gene-3 (LAG-3), T cell immunoglobulin and mucin-domain containing-3 (TIM-3), T cell immunoglobulin and ITIM domain (TIGIT), V-domain Ig suppressor of T cell activation (VISTA), and so on. The investigations about these molecules have generated promising results in preclinical studies and/or clinical trials. In this review, we discussed the structure and expression of these newly-characterized immune checkpoints molecules, presented the current progress and understanding of them. Moreover, we summarized the clinical data pertinent to these recent immune checkpoint molecules as well as their application prospects.
Collapse
Affiliation(s)
- Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Linping Xu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China. .,Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| | - Suxia Luo
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
69
|
C Guven D, Kilickap S, Guner G, Taban H, Dizdar O. Development of de novo psoriasis during nivolumab therapy in a patient with small cell lung cancer. J Oncol Pharm Pract 2019; 26:256-258. [PMID: 31566114 DOI: 10.1177/1078155219877234] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION The immune checkpoint inhibitors recently entered to small cell lung cancer (SCLC) stage, firstly in the third and recently in the first lines of therapy. This efficacy comes at the expense of many toxicities including skin toxicity. This toxicity is usually in the form of rash and pruritis; however, rare reactions like psoriasis can also be seen. CASE REPORT Herein, we report an SCLC case who developed de novo psoriasis while treated with nivolumab as the third-line treatment for SCLC. MANAGEMENT AND OUTCOME The psoriatic plaques were regressed with the topical highly potent steroid therapy, and immunotherapy was continued without further complications. DISCUSSION We think that rare adverse events like de novo psoriasis are important considering the expanding role of these agents; their timely recognition and treatment are important in the management of cancer patients.
Collapse
Affiliation(s)
- Deniz C Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Saadettin Kilickap
- Department of Preventive Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Gurkan Guner
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Hakan Taban
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Omer Dizdar
- Department of Preventive Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
70
|
Zarrabi K, Paroya A, Wu S. Emerging therapeutic agents for genitourinary cancers. J Hematol Oncol 2019; 12:89. [PMID: 31484560 PMCID: PMC6727406 DOI: 10.1186/s13045-019-0780-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 08/21/2019] [Indexed: 12/11/2022] Open
Abstract
The treatment of genitourinary malignancies has dramatically evolved over recent years. Renal cell carcinoma, urothelial carcinoma of the bladder, and prostate adenocarcinoma are the most commonly encountered genitourinary malignancies and represent a heterogeneous population of cancers, in both histology and approach to treatment. However, all three cancers have undergone paradigm shifts in their respective therapeutic landscapes due to a greater understanding of their underlying molecular mechanisms and oncogenic drivers. The advance that has gained the most recent traction has been the advent of immunotherapies, particularly immune checkpoint inhibitors. Immunotherapy has increased overall survival and even provided durable responses in the metastatic setting in some patients. The early success of immune checkpoint inhibitors has led to further drug development with the emergence of novel agents which modulate the immune system within the tumor microenvironment. Notwithstanding immunotherapy, investigators are also developing novel agents tailored to a variety of targets including small-molecule tyrosine kinase inhibitors, mTOR inhibitors, and novel fusion proteins to name a few. Erdafitinib has become the first targeted therapy approved for metastatic bladder cancer. Moreover, the combination therapy of immune checkpoint inhibitors with targeted agents such as pembrolizumab or avelumab with axitinib has demonstrated both safety and efficacy and just received FDA approval for their use. We are in an era of rapid progression in drug development with multiple exciting trials and ongoing pre-clinical studies. We highlight many of the promising new emerging therapies that will likely continue to improve outcomes in patients with genitourinary malignancies.
Collapse
Affiliation(s)
- Kevin Zarrabi
- Department of Medicine, Stony Brook University Hospital, 9447 SUNY, Stony Brook, NY 11794-9447 USA
| | - Azzam Paroya
- Department of Medicine, Stony Brook University Hospital, 9447 SUNY, Stony Brook, NY 11794-9447 USA
| | - Shenhong Wu
- Department of Medicine, Stony Brook University Hospital, 9447 SUNY, Stony Brook, NY 11794-9447 USA
- Division of Hematology/Oncology, Department of Medicine, Northport VA Medical Center, Northport, NY USA
| |
Collapse
|
71
|
Pierantoni F, Maruzzo M, Gardi M, Bezzon E, Gardiman MP, Porreca A, Basso U, Zagonel V. Immunotherapy and urothelial carcinoma: An overview and future prospectives. Crit Rev Oncol Hematol 2019; 143:46-55. [PMID: 31476551 DOI: 10.1016/j.critrevonc.2019.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 08/06/2019] [Accepted: 08/22/2019] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Urothelial carcinoma (UC) is a common malignancy with a high mortality rate when metastatic. Traditionally, systemic therapy consisted in platinum-based regimens as first-line, with Taxanes or Vinflunine as further lines. Recently, checkpoint inhibitors (CPIs) immunotherapy has emerged as a new therapeutic option. METHODS We searched in Medline, Pubmed and ClinicalTrial.gov databases for the relevant literature, reviewing the results of published trials and the design of ongoing studies involving CPIs in UC. RESULT Strong evidence supports the use of CPIs after failure of Cisplatin-based chemotherapy, although no predictive parameter is available so far. Expression of Programmed-Death-1-Ligand has given conflicting results, and is currently indicated only for the selection of Cisplatin-ineligible patients who should receive CPIs. CONCLUSION The therapeutic landscape of UC is rapidly changing due to the availability of CPIs. Neoadjuvant trials with CPIs and trials combining two CPIs are promising and will further expand the use of immunotherapy.
Collapse
Affiliation(s)
- Francesco Pierantoni
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCSS, Padua, Italy.
| | - Marco Maruzzo
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCSS, Padua, Italy
| | - Mario Gardi
- Urology Unit, Department of Surgery, Sant'Antonio Hospital, Padua, Italy
| | - Elisabetta Bezzon
- Radiology Unit, Department of Imaging and Medical Physics, Istituto Oncologico Veneto IOV IRCSS, Padua, Italy
| | - Marina Paola Gardiman
- Surgical Pathology and Cytopathology Unit, Department of Medicine, University Hospital of Padua, Italy
| | - Angelo Porreca
- Urology Unit, Policlinico Abano Terme, Abano Terme, Italy
| | - Umberto Basso
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCSS, Padua, Italy
| | - Vittorina Zagonel
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCSS, Padua, Italy
| |
Collapse
|
72
|
Najberg M, Haji Mansor M, Boury F, Alvarez-Lorenzo C, Garcion E. Reversing the Tumor Target: Establishment of a Tumor Trap. Front Pharmacol 2019; 10:887. [PMID: 31456685 PMCID: PMC6699082 DOI: 10.3389/fphar.2019.00887] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/15/2019] [Indexed: 12/19/2022] Open
Abstract
Despite the tremendous progress made in the field of cancer therapy in recent years, certain solid tumors still cannot be successfully treated. Alongside classical treatments in the form of chemotherapy and/or radiotherapy, targeted treatments such as immunotherapy that cause fewer side effects emerge as new options in the clinics. However, these alternative treatments may not be useful for treating all types of cancers, especially for killing infiltrative and circulating tumor cells (CTCs). Recent advances pursue the trapping of these cancer cells within a confined area to facilitate their removal for therapeutic and diagnostic purposes. A good understanding of the mechanisms behind tumor cell migration may drive the design of traps that mimic natural tumor niches and guide the movement of the cancer cells. To bring this trapping idea into reality, strong efforts are being made to create structured materials that imitate myelinated fibers, blood vessels, or pre-metastatic niches and incorporate chemical cues such as chemoattractants or adhesive proteins. In this review, the different strategies used (or could be used) to trap tumor cells are described, and relevant examples of their performance are analyzed.
Collapse
Affiliation(s)
- Mathie Najberg
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R + D Pharma Group (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Muhammad Haji Mansor
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
- Center for Education and Research on Macromolecules (CERM), Université de Liège, Liège, Belgium
| | - Frank Boury
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R + D Pharma Group (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Emmanuel Garcion
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
| |
Collapse
|
73
|
Liu D, Zhao J, Song Y. Engineering switchable and programmable universal CARs for CAR T therapy. J Hematol Oncol 2019; 12:69. [PMID: 31272471 PMCID: PMC6610960 DOI: 10.1186/s13045-019-0763-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 06/25/2019] [Indexed: 12/27/2022] Open
Abstract
A traditional chimeric antigen receptor (CAR) has a fixed design, and one type of CAR T cells can only target one antigen epitope. This rigid design limits clinical application and leads to exceptionally high manufacturing cost. New CARs are being engineered with a modular approach so that the antigen recognition domain is split from the signaling domain of a conventional CAR, hence the target antigen can be switched or re-directed more readily without the requirement of re-engineering the CAR T cells. This CAR system can therefore serve as a universal CAR (UniCAR). The UniCAR platform has a modular design that splits the conventional CAR to 2 separate components: (1) a signaling module that binds to a specific epitope on the switching molecule and (2) a switching module with an antigen-binding domain and a switching epitope specifically recognized by the signaling module. A variety of switchable CARs have been engineered. The switchable modular designs include the dimerizing platforms using leucine zippers and biotin-avidin system, and the neo-epitope tagging platform using FITC, 5B9, and PNE. The switch molecule serves as a synapse between the CAR T cells and the target tumor cells. The universal CAR platforms are highly versatile, are easily re-programmable, and therefore have a vast potential for broad application and may significantly lower the cost of CAR T cell therapy. However, the current modular design of the switching molecules relies on adding exogenous sequences/epitopes. These unnatural epitopes can potentially lead to new antigenicity which may lead to generation of blocking antibodies. Furthermore, the generation, preparation, and clinical applications of the switching modules per se may involve additional clinical trials and regulatory examination for safety and efficacy, since repeated administrations of these molecules/"drugs" are anticipated. Thus, these switching molecules and UniCAR CAR T cells may require separate clinical trials and invoke different regulatory processes. This whole field is medically appealing and could present new challenges in the development of novel immunotherapeutic agents.
Collapse
Affiliation(s)
- Delong Liu
- Department of Oncology, The First affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY USA
| | - Juanjuan Zhao
- The affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| | - Yongping Song
- The affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| |
Collapse
|
74
|
Wang Q, Yang S, Wang K, Sun SY. MET inhibitors for targeted therapy of EGFR TKI-resistant lung cancer. J Hematol Oncol 2019; 12:63. [PMID: 31227004 PMCID: PMC6588884 DOI: 10.1186/s13045-019-0759-9] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/14/2019] [Indexed: 01/10/2023] Open
Abstract
Treatment of non-small cell lung cancer (NSCLC) harboring epidermal growth factor receptor (EGFR) activating mutation with EGFR-TKIs has achieved great success, yet faces the development of acquired resistance as the major obstacle to long-term disease remission in the clinic. MET (or c-MET) gene amplification has long been known as an important resistance mechanism to first- or second-generation EGFR-TKIs in addition to the appearance of T790 M mutation. Recent preclinical and clinical studies have suggested that MET amplification and/or protein hyperactivation is likely to be a key mechanism underlying acquired resistance to third-generation EGFR-TKIs such as osimertinib as well, particularly when used as a first-line therapy. EGFR-mutant NSCLCs that have relapsed from first-generation EGFR-TKI treatment and have MET amplification and/or protein hyperactivation should be insensitive to osimertinib monotherapy. Therefore, combinatorial therapy with osimertinib and a MET or even a MEK inhibitor should be considered for these patients with resistant NSCLC carrying MET amplification and/or protein hyperactivation.
Collapse
Affiliation(s)
- Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China.
| | - Sen Yang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Kai Wang
- Department of Respiratory Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, School of Medicine and Winship Cancer Institute, Emory University, 1365-C Clifton Road, C3088, Atlanta, GA, 30322, USA.
| |
Collapse
|
75
|
Zhang Y, Xu L, Chen S, Zha X, Wei W, Li Y. Identification of TCR Vβ11-2- Dβ1- Jβ1-1 T cell clone specific for WT1 peptides using high-throughput TCRβ gene sequencing. Biomark Res 2019; 7:12. [PMID: 31223481 PMCID: PMC6570921 DOI: 10.1186/s40364-019-0163-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023] Open
Abstract
We previously identified a TCR Vβ21 T cell clone which was specific to CML patients, and demonstrated that TCR Vα13/β21 gene-modified CD3+ T cells had specific cytotoxicity for HLA-A11+ K562 cells. However, it remains unclear which antigen is specifically recognized by the TCR Vβ21 T cell clone. In this study, CD3+ T cells from healthy donor peripheral blood were stimulated with the WT1 peptide or mixed BCR-ABL peptides in the presence or absence of IL-2 and IL-7. The distribution of the TCR Vβ repertoire was analyzed after different stimulations. We found that the mixed BCR-ABL peptides induced clonally expanded Vβ7-9-Dβ2-Jβ2-7 T cells while the Wilms Tumor 1 peptide induced clonally expanded Vβ11-2-Dβ1-Jβ1-1 T cells by high-throughput TCRβ sequencing and GeneScan. Interestingly, the sequence and CDR3 motif of Vβ11-2 T cell clone are similar to the TCR Vβ21 (a different TCR V region naming system) T cell clone that we previously found in CML patients. Thus, our findings suggest that the TCR Vβ21 T cell clone found in CML patients might be a T cell clone that specifically recognizes WT1.
Collapse
Affiliation(s)
- Yikai Zhang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, 601 Huang Pu Da Dao Xi, 510632 Guangzhou, People’s Republic of China
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632 China
| | - Ling Xu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, 601 Huang Pu Da Dao Xi, 510632 Guangzhou, People’s Republic of China
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632 China
| | - Shaohua Chen
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, 601 Huang Pu Da Dao Xi, 510632 Guangzhou, People’s Republic of China
| | - Xianfeng Zha
- Department of Clinical Laboratory, First Affiliated Hospital, Jinan University, Guangzhou, 510632 China
| | - Wei Wei
- Guangzhou Municipality Tianhe Nuoya Bio-engineering Co. Ltd, Guangzhou, 510663 China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, 601 Huang Pu Da Dao Xi, 510632 Guangzhou, People’s Republic of China
| |
Collapse
|
76
|
Appiya Santharam M, Khan FU, Naveed M, Ali U, Ahsan MZ, Khongorzul P, Shoaib RM, Ihsan AU. Interventions to chronic prostatitis/Chronic pelvic pain syndrome treatment. Where are we standing and what's next? Eur J Pharmacol 2019; 857:172429. [PMID: 31170381 DOI: 10.1016/j.ejphar.2019.172429] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/29/2022]
Abstract
Chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) is a frustrating syndrome. The pathogenesis and state of the art treatment of CP/CPPS are not known. A wide variety of therapies including anti-inflammatories, antibiotics, alpha-blockers, neuropathic pain modulators, and 5α-reductase inhibitors are in practice. These treatment strategies focus on alleviating symptoms in specific domains without treating root-cause and therapeutic outcome is far from satisfactory. We review the literature on current pharmacological treatments for CP/CPPS in detail and suggest future perspectives to modify the treatment strategies. We suggest that introducing novel treatment strategies such as gene editing, and Tregs expressing chimeric receptors may improve the treatment outcomes by inducing immune tolerance and controlling expression of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Madanraj Appiya Santharam
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester LE1 9HN, United Kingdom
| | - Farhan Ullah Khan
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, 211198, PR China; Shanghai Jiao Tong University, School of Pharmacy, 800 Dongchuan Road, Shanghai, 200240, China
| | - Muhammad Naveed
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Jiangsu Province, Nanjing, 211166, PR China
| | - Usman Ali
- Shanghai Jiao Tong University, School of Pharmacy, 800 Dongchuan Road, Shanghai, 200240, China
| | - Muhammad Zaeem Ahsan
- Shanghai Jiao Tong University, School of Pharmacy, 800 Dongchuan Road, Shanghai, 200240, China
| | - Puregmaa Khongorzul
- State Key Laboratory of Natural Medicines, School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Rana Muhammad Shoaib
- Shanghai Jiao Tong University, School of Pharmacy, 800 Dongchuan Road, Shanghai, 200240, China
| | - Awais Ullah Ihsan
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu Province, Nanjing, 211198, PR China.
| |
Collapse
|
77
|
Thang DC, Wang Z, Lu X, Xing B. Precise cell behaviors manipulation through light-responsive nano-regulators: recent advance and perspective. Theranostics 2019; 9:3308-3340. [PMID: 31244956 PMCID: PMC6567964 DOI: 10.7150/thno.33888] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023] Open
Abstract
Nanotechnology-assisted spatiotemporal manipulation of biological events holds great promise in advancing the practice of precision medicine in healthcare systems. The progress in internal and/or external stimuli-responsive nanoplatforms for highly specific cellular regulations and theranostic controls offer potential clinical translations of the revolutionized nanomedicine. To successfully implement this new paradigm, the emerging light-responsive nanoregulators with unparalleled precise cell functions manipulation have gained intensive attention, providing UV-Vis light-triggered photocleavage or photoisomerization studies, as well as near-infrared (NIR) light-mediated deep-tissue applications for stimulating cellular signal cascades and treatment of mortal diseases. This review discusses current developments of light-activatable nanoplatforms for modulations of various cellular events including neuromodulations, stem cell monitoring, immunomanipulation, cancer therapy, and other biological target intervention. In summary, the propagation of light-controlled nanomedicine would place a bright prospect for future medicine.
Collapse
Affiliation(s)
- Do Cong Thang
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| | - Zhimin Wang
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| | - Xiaoling Lu
- International Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Bengang Xing
- Sino-Singapore International Joint Research Institute (SSIJRI), Guangzhou 510000, China
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| |
Collapse
|
78
|
Abstract
Currently, chemotherapy remains the standard treatment for first- and second-line management of small cell lung cancer (SCLC). Immunotherapy has made progress in the treatment of SCLC, and nivolumab, pembrolizumab, atezolizumab, and durvalumab have led to significant improvements in clinical outcomes of SCLC. Regarding options in other classes of therapy, the cytotoxic drug lurbinectedin was granted orphan drug status based on a remarkable objective response rate of 39.3%. In addition, an increase in progression-free survival (PFS) was achieved in a phase II study of anlotinib (ALTER 1202). Future prospects for even better outcomes in SCLC lie in novel ways to integrate immunotherapy and small-molecule TKI drugs. Innovative clinical trial designs are needed to efficiently explore the increasing number of options with new drugs and new combinations thereof for SCLC.
Collapse
Affiliation(s)
- Sen Yang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Zhe Zhang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
79
|
Kong Y, Jia B, Zhao C, Claxton DF, Sharma A, Annageldiyev C, Fotos JS, Zeng H, Paulson RF, Prabhu KS, Zheng H. Downregulation of CD73 associates with T cell exhaustion in AML patients. J Hematol Oncol 2019; 12:40. [PMID: 31014364 PMCID: PMC6480867 DOI: 10.1186/s13045-019-0728-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 03/31/2019] [Indexed: 12/25/2022] Open
Abstract
Background Successful treatment for acute myeloid leukemia (AML) remains challenging. Inhibiting immune checkpoint to enhance anti-tumor response is an attractive strategy for effective leukemia therapeutics. CD73 is a recently recognized immune checkpoint mediator that is highly expressed on tumor cells and stromal cells in tumor microenvironment. The ectonucleotidase activity of CD73 catalyzes AMP to adenosine, which subsequently inhibits anti-tumor immune responses. In this study, we aim to explore the effect of CD73 in AML. Methods Peripheral blood samples collected from patients with newly diagnosed AML (n = 27) were used in this study. CD73 expression on each immune cell component was examined by flow cytometry. Phenotypic study of CD73-expressing T cells and analysis of the correlation between CD73 and other immune checkpoints were performed using flow cytometry-based assays. Functional status of CD73+ vs. CD73− T cells was assessed in an in vitro cytokine release assay upon CD3/CD28 antibody stimulation. Results In contrast to the long recognized immune suppressive effect of CD73-adenosine signaling in tumor tissue, we made a striking observation that in AML, CD73 expression on CD8 T cells associates with an increased immune response. CD73+ CD8 T cells are more functional, whereas CD73− CD8 T cells exhibit features of exhaustion manifested by high expression of inhibitory receptors such as PD-1 and TIGIT, increased intracellular expression of Eomes, reduced capacity of cytokine production, and high susceptibility to apoptosis. Conclusions Our data highlight the potential of CD73 as a double-edged sword in anti-leukemia immunity and argue strongly for the combinational treatment by adding immune checkpoint inhibitors to the CD73-targeting approaches.
Collapse
Affiliation(s)
- Yaxian Kong
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA.,Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Department of Intensive Care Unit, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Bei Jia
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Chenchen Zhao
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - David F Claxton
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Arati Sharma
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA.,Department of Pharmacology, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Charyguly Annageldiyev
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Joseph S Fotos
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Hui Zeng
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Robert F Paulson
- Department of Veterinary and Biomedical Sciences, Penn State University College of Agricultural Sciences, University Park, Harrisburg, PA, 16802, USA
| | - K Sandeep Prabhu
- Department of Veterinary and Biomedical Sciences, Penn State University College of Agricultural Sciences, University Park, Harrisburg, PA, 16802, USA
| | - Hong Zheng
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
80
|
Gao Y, Shi S, Ma W, Chen J, Cai Y, Ge L, Li L, Wu J, Tian J. Bibliometric analysis of global research on PD-1 and PD-L1 in the field of cancer. Int Immunopharmacol 2019; 72:374-384. [PMID: 31030093 DOI: 10.1016/j.intimp.2019.03.045] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/06/2019] [Accepted: 03/22/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To identify the cooperation of authors, countries, and institutions and explore the hot topics and future prospects regarding programmed cell death protein 1 (PD-1) and programmed cell death 1 ligand 1 (PD-L1) research. MATERIALS AND METHODS Publications on PD-1 and PD-L1 research were retrieved from the Web of Science Core Collection. Bibliometric analyses were performed using VOSviewer 1.6.9, HistCite 2.1, and CiteSpace V software. Network maps were generated to evaluate the collaborations between different authors, countries, and institutions. RESULTS A total of 7359 articles related to PD-1 and PD-L1 research in the field of cancer were identified. We observed rapid growth in the number of publications since 2014. Oncoimmunology (326, 4.43%) had the highest number of publications, while N Engl J Med (18,828 co-citations, 6.30%) was the most co-cited journal. Hodi FS and Wolchok JD were key researchers. There were active collaborations among the top authors. The USA was the leading contributor in this field with 3496 publications. Active cooperations between countries and between institutions were observed. The main hot topics included matters related to the efficacy and prognosis of various tumors after treatment with PD-1 and/or PD-L1 inhibitors, as well as the activation or expression of important immunological factors targeted at tumors. CONCLUSION PD-1 and PD-L1 studies have significantly increased after 2014. The USA contributed the most publications. There were active cooperations between authors, countries, and institutions. Further research should expand and develop new topics such as those likely to boost therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Ya Gao
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Shuzhen Shi
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Wenjuan Ma
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Ji Chen
- Evidence-Based Nursery Center, School of Nursing, Lanzhou University, Lanzhou, China
| | - Yitong Cai
- Evidence-Based Nursery Center, School of Nursing, Lanzhou University, Lanzhou, China
| | - Long Ge
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China; School of Public Health, Lanzhou University, Lanzhou, China
| | - Lun Li
- Shanghai Cancer Center of Fudan University, Shanghai, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jinhui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China.
| |
Collapse
|
81
|
Aujla A, Aujla R, Liu D. Inotuzumab ozogamicin in clinical development for acute lymphoblastic leukemia and non-Hodgkin lymphoma. Biomark Res 2019; 7:9. [PMID: 31011424 PMCID: PMC6458768 DOI: 10.1186/s40364-019-0160-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/27/2019] [Indexed: 12/26/2022] Open
Abstract
B cell acute lymphoblastic leukemia (ALL) and non-Hodgkin lymphoma (NHL) frequently express CD19, CD20 and CD22 on the cell surfaces. Immunotherapeutic agents including antibodies and chimeric antigen receptor T cells are widely studied in clinical trials. Several antibody-drug conjugates (ADC) have been approved for clinical use (gemtuzumab ozogamicin in acute myeloid leukemia and brentuximab vedotin in Hodgkin lymphoma as well as CD30+ anaplastic large cell lymphoma). Inotuzumab ozogamicin (INO), a CD22 antibody conjugated with calicheamicin is one of the newest ADCs. INO has been approved for treatment of relapsed /refractory B cell precursor ALL. Multiple ongoing trials are evaluating its role in the relapsed /refractory B cell NHL. This review summarized recent development in INO applications for ALL and NHL.
Collapse
Affiliation(s)
- Amandeep Aujla
- 1Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595 USA
| | - Ravijot Aujla
- 2Punjab Institute of Medical Sciences, Jalandhar, Punjab 144006 India
| | - Delong Liu
- 1Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595 USA.,3Department of Oncology, The First affiliated hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
82
|
PEST-containing nuclear protein regulates cell proliferation, migration, and invasion in lung adenocarcinoma. Oncogenesis 2019; 8:22. [PMID: 30872582 PMCID: PMC6418141 DOI: 10.1038/s41389-019-0132-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/08/2019] [Accepted: 02/25/2019] [Indexed: 12/23/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. PEST-containing nuclear protein (PCNP) has been found in the nucleus of cancer cells. Whether PCNP plays a role in the growth of lung adenocarcinoma is still unknown. In the present study, the results indicated that the level of PCNP in lung adenocarcinoma tissue was significantly higher than that in corresponding adjacent non-tumor tissue. Over-expression of PCNP promoted the proliferation, migration, and invasion of lung adenocarcinoma cells, while down-regulation of PCNP exhibited opposite effects. PCNP over-expression decreased apoptosis through up-regulating the expression levels of phospho (p)-signal transducers and activators of transcription (STAT) 3 and p-STAT5 in lung adenocarcinoma cells, whereas PCNP knockdown showed opposite trends. PCNP overexpression enhanced autophagy by increasing the expression levels of p-phosphatidylinositol 3-kinase (PI3K), p-Akt, and p-mammalian target of rapamycin (mTOR) in lung adenocarcinoma cells, however an opposite trend was observed in the sh-PCNP group. In addition, overexpression of PCNP showed the tumor-promoting effect on xenografted lung adenocarcinoma, while PCNP knockdown reduced the growth of lung adenocarcinoma via regulating angiogenesis. Our study elucidates that PCNP can regulate the procession of human lung adenocarcinoma cells via STAT3/5 and PI3K/Akt/mTOR signaling pathways. PCNP may be considered as a promising biomarker for the diagnosis and prognosis in patients with lung adenocarcinoma. Furthermore, PCNP can be a novel therapeutic target and potent PCNP inhibitors can be designed and developed in the treatment of lung adenocarcinoma.
Collapse
|
83
|
Qin S, Li A, Yi M, Yu S, Zhang M, Wu K. Recent advances on anti-angiogenesis receptor tyrosine kinase inhibitors in cancer therapy. J Hematol Oncol 2019; 12:27. [PMID: 30866992 PMCID: PMC6417086 DOI: 10.1186/s13045-019-0718-5] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/05/2019] [Indexed: 02/08/2023] Open
Abstract
Angiogenesis has always been the topic of major scientific interest in the field of malignant tumors. Nowadays, targeting angiogenesis has achieved success in various carcinomas by several mechanisms, including the use of anti-angiogenic small molecule receptor tyrosine kinase inhibitors (TKIs). The development of TKIs targeting pro-angiogenic receptors, mainly vascular endothelial growth factor receptor (VEGFR) family, have significantly improved the outcome of certain types of cancers, like renal cell carcinoma, hepatocellular carcinoma, and colorectal carcinoma. However, the general response rate is not very satisfactory. The particular toxicity profile and resistance to anti-angiogenic targeted agents are unavoidable, and no specific marker is available to screen responsive patients to TKIs for precision therapy. To date, about 11 anti-angiogenic TKIs with different binding capacities to angiogenic receptor tyrosine kinase have been approved for the treatment of patients with advanced cancers. This review presents all approved anti-angiogenic small molecule receptor TKIs so far with an emphasis on their indications and clinical efficacy. We also discuss the combination between TKIs and immune checkpoint blockade inhibitors based on the most recent exciting outcome in immunotherapy.
Collapse
Affiliation(s)
- Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Anping Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Mingsheng Zhang
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China. .,Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
84
|
Zhao J, Song Y, Liu D. Clinical trials of dual-target CAR T cells, donor-derived CAR T cells, and universal CAR T cells for acute lymphoid leukemia. J Hematol Oncol 2019; 12:17. [PMID: 30764841 PMCID: PMC6376657 DOI: 10.1186/s13045-019-0705-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 02/07/2019] [Indexed: 02/08/2023] Open
Abstract
The current treatment for pediatric acute lymphoblastic leukemia (ALL) is highly successful with high cure rate. However, the treatment of adult ALL remains a challenge, particularly for refractory and/or relapsed (R/R) ALL. The advent of new targeted agents, blinatumomab, inotuzumab ozogamycin, and chimeric antigen receptor (CAR) T cells, are changing the treatment paradigm for ALL. Tisagenlecleucel (kymriah, Novartis) is an autologous CD19-targeted CAR T cell product approved for treatment of R/R B cell ALL and lymphoma. In an attempt to reduce the relapse rate and treat those relapsed patients with antigen loss, donor-derived CAR T cells and CD19/CD22 dual-target CAR T cells are in clinical trials. Gene-edited “off-the-shelf” universal CAR T cells are also undergoing active clinical development. This review summarized new clinical trials and latest updates at the 2018 ASH Annual Meeting on CAR T therapy for ALL with a focus on dual-target CAR T and universal CAR T cell trials.
Collapse
Affiliation(s)
- Juanjuan Zhao
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008, China
| | - Yongping Song
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008, China
| | - Delong Liu
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008, China.
| |
Collapse
|
85
|
Prabhash K, Abraham G, Menon N, Patil V, Joshi A. The efficacy of low-dose immunotherapy in head-and-neck cancer. CANCER RESEARCH, STATISTICS, AND TREATMENT 2019. [DOI: 10.4103/crst.crst_102_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
86
|
Yang H, Wu M, Shen Y, Lei T, Mi L, Leng X, Ping L, Xie Y, Song Y, Cen X, Zhu J. Treatment Strategies and Prognostic Factors of Primary Gastric Diffuse Large B Cell Lymphoma: A Retrospective Multicenter Study of 272 Cases from the China Lymphoma Patient Registry. Int J Med Sci 2019; 16:1023-1031. [PMID: 31341416 PMCID: PMC6643119 DOI: 10.7150/ijms.34175] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 05/08/2019] [Indexed: 01/28/2023] Open
Abstract
Background: The respective and combinatorial roles of surgery, Rituximab and chemotherapy in primary gastric diffuse large B cell lymphoma (PGDLBCL) therapy remained unclear. The purpose of the study was to evaluate present treatment strategies and prognostic factors of PGDLBCL. Methods: 272 cases (from 1994-1 to 2015-12) were retrospectively analyzed. According to the therapy regimen, patients were classified into four groups: chemotherapy (C), chemotherapy + surgery (C+S), Rituximab + chemotherapy (R+C), and Rituximab + chemotherapy + surgery (R+C+S). Results: The 3-year progression-free survival (PFS) and 3-year overall survivals (OS) of the entire cohort were 77.0% and 81.2% respectively (median follow-up time: 44.3 months). Survival of surgery-treated patients was superior to the survival of those receiving drug therapy alone (PFS: 82.6% vs. 74.7%, p=0.015; OS: 87.8% vs. 78.6%, p=0.036). Rituximab showed significant clinical benefit in OS (87.1% vs. 75.0%, p=0.007), especially in advanced-stage or high risk (IPI 3-5) patients. Group C had the lowest PFS and OS among the four groups, while the survival of other three groups were similar (Group C vs. Group C+S vs. Group R+C vs. Group R+C+S: 3-year PFS: 67.2% vs. 81.4% vs. 81.2% vs. 81.8%, p=0.002; 3-year OS: 68.4% vs. 85.4% vs. 87.2% vs. 88.6%, p<0.001). Multivariate analysis showed that IPI and therapy regimens were highly predictive for both PFS and OS. Conclusions: Our results suggested that the combinations of chemotherapy and surgery, or chemotherapy and Rituximab, are superior to other treatment strategies for PGDLBCL. IPI and therapy regimens are independent predictors of outcomes. Future prospective trial is warranted.
Collapse
Affiliation(s)
- Haiyan Yang
- Department of Lymphoma, Zhejiang Cancer Hospital, Postal address: No. 1, Bansan Road, Hangzhou, Zhejiang, China
| | - Meng Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Postal address: No 52, Fucheng Road, Haidian District, Beijing, China. 100142
| | - Ye Shen
- Department of Hematology, Peking University First Hospital, Postal address: No. 8, Xishiku Street, Xicheng District, Beijing, China. 100034
| | - Tao Lei
- Department of Lymphoma, Zhejiang Cancer Hospital, Postal address: No. 1, Bansan Road, Hangzhou, Zhejiang, China
| | - Lan Mi
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of outreach and industrial Affairs, Peking University Cancer Hospital & Institute, Postal address: No 52, Fucheng Road, Haidian District, Beijing, China. 100142
| | - Xin Leng
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Postal address: No 52, Fucheng Road, Haidian District, Beijing, China. 100142
| | - Lingyan Ping
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Postal address: No 52, Fucheng Road, Haidian District, Beijing, China. 100142
| | - Yan Xie
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Postal address: No 52, Fucheng Road, Haidian District, Beijing, China. 100142
| | - Yuqin Song
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Postal address: No 52, Fucheng Road, Haidian District, Beijing, China. 100142
| | - Xinan Cen
- Department of Hematology, Peking University First Hospital, Postal address: No. 8, Xishiku Street, Xicheng District, Beijing, China. 100034
| | - Jun Zhu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Postal address: No 52, Fucheng Road, Haidian District, Beijing, China. 100142
| |
Collapse
|