1
|
Li N, Zhan X. Integrated genomic analysis of proteasome alterations across 11,057 patients with 33 cancer types: clinically relevant outcomes in framework of 3P medicine. EPMA J 2021; 12:605-627. [PMID: 34956426 DOI: 10.1007/s13167-021-00256-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 09/16/2021] [Indexed: 12/30/2022]
Abstract
Relevance Proteasome, a cylindrical complex containing 19S regulatory particle lid, 19S regulatory particle base, and 20S core particle, acted as a major mechanism to regulate the levels of intracellular proteins and degrade misfolded proteins, which involved in many cellular processes, and played important roles in cancer biological processes. Elucidation of proteasome alterations across multiple cancer types will directly contribute to cancer medical services in the context of predictive, preventive, and personalized medicine (PPPM / 3P medicine). Purpose This study aimed to investigate proteasome gene alterations across 33 cancer types for discovery of effective biomarkers and therapeutic targets in the framework of PPPM practice in cancers. Methods Proteasome gene data, including gene expression RNAseq, somatic mutation, tumor mutation burden (TMB), copy number variant (CNV), microsatellite instability (MSI) score, clinical characteristics, immune phenotype, 22 immune cells, cancer stemness index, drug sensitivity, and related pathways, were systematically analyzed with publically available database and bioinformatics across 11,057 patients with 33 cancer types. Results Differentially expressed proteasome genes were extensively found between tumor and control tissues. PSMB4 occurred the top mutation event among proteasome genes, and those proteasome genes were significantly associated with TMB and MSI score. Most of proteasome genes were positively related to CNV among single deletion, control copy number, and single gain. Kaplan-Meier curves and COX regression survival analysis showed proteasome genes were significantly associated with patient survival rate across 33 cancer types. Furthermore, the expressions of proteasome genes were significantly different among different clinical stages and immune subtypes. The expressions of proteasome genes were correlated with immune-related scores (ImmuneScore, StromalScore, and ESTIMATEScore), 22 immune cells, and cancer stemness. The sensitivities of multiple drugs were closely related to proteasome gene expressions. The identified proteasome and proteasome-interacted proteins were significantly enriched in various cancer-related pathways. Conclusions This study provided the first landscape of proteasome alterations across 11,057 patients with 33 cancer types and revealed that proteasome played a significant and wide functional role in cancer biological processes. These findings are the precious scientific data to reveal the common and specific alterations of proteasome genes among 33 cancer types, which benefits the research and practice of PPPM in cancers. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-021-00256-z.
Collapse
Affiliation(s)
- Na Li
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, 440 Jiyan Road, Jinan, Shandong 250117 People's Republic of China.,Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan, Shandong 250117 People's Republic of China
| | - Xianquan Zhan
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, 440 Jiyan Road, Jinan, Shandong 250117 People's Republic of China.,Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan, Shandong 250117 People's Republic of China.,Gastroenterology Research Institute and Clinical Center, Shandong First Medical University, 38 Wuying Shan Road, Jinan, Shandong 250031 People's Republic of China
| |
Collapse
|
2
|
Toress-Collado AX, Nazarian R, Jazirehi AR. Rescue of cell cycle progression in BRAF V600E inhibitor-resistant human melanoma by a chromatin modifier. Tumour Biol 2017; 39:1010428317721620. [PMID: 28936920 DOI: 10.1177/1010428317721620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The BRAFV600E-specific inhibitor vemurafenib blocks mitogen-activated protein kinase pathway and induces cell cycle arrest at G0/G1 phase leading to apoptosis of melanomas. To gain an understanding of the dynamics of cell cycle regulation during vemurafenib therapy, we analyzed several vemurafenib-resistant human melanoma sublines derived from BRAFV600E harboring vemurafenib-sensitive parental lines. Vemurafenib provoked G0/G1 phase arrest in parental but not in vemurafenib-resistant sublines. We hypothesized that refractoriness of vemurafenib-resistant sublines to vemurafenib-mediated cell cycle inhibition can be partially rescued by the chromatin modifier suberoylanilide hydroxamic acid. Suberoylanilide hydroxamic acid promoted G2/M arrest at expense of S phase irrespective of vemurafenib sensitivity. In parental lines, combination of suberoylanilide hydroxamic acid and vemurafenib induced both G0/G1 arrest and apoptosis, whereas in vemurafenib-resistant sublines combination induced G0/G1 as well as G2/M arrest resulting in dramatic cytostasis. Vemurafenib-resistant sublines exhibited extracellular signal-regulated protein kinases 1 and 2 but not AKT and hyperphosphorylation. Gene expression profiling revealed mitogen-activated protein kinase hyperactivation and deregulations of cyclins and cyclin-dependent kinases in vemurafenib-resistant sublines, all of which were reversed by suberoylanilide hydroxamic acid; changes that may explain the cytostatic effects of suberoylanilide hydroxamic acid. These results suggest that unresponsiveness of vemurafenib-resistant sublines to the biological effects of vemurafenib may be amenable by suberoylanilide hydroxamic acid. These in vitro results, while require further investigation, may provide rational biological basis for combination therapy in the management of vemurafenib-resistant melanoma.
Collapse
Affiliation(s)
- Antoni X Toress-Collado
- 1 Division of Surgical Oncology, Department of Surgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ramin Nazarian
- 2 Division of Hematology/Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,3 Jonsson Comprehensive Cancer Center (JCCC), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ali R Jazirehi
- 1 Division of Surgical Oncology, Department of Surgery, University of California, Los Angeles, Los Angeles, CA, USA.,3 Jonsson Comprehensive Cancer Center (JCCC), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
3
|
Retraction: Proteasome Inhibition Blocks NF-κB and ERK1/2 Pathways, Restores Antigen Expression, and Sensitizes Resistant Human Melanoma to TCR-Engineered CTLs. Mol Cancer Ther 2017; 16:977. [DOI: 10.1158/1535-7163.mct-17-0084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
4
|
Singha B, Gatla HR, Phyo S, Patel A, Chen ZS, Vancurova I. IKK inhibition increases bortezomib effectiveness in ovarian cancer. Oncotarget 2016; 6:26347-58. [PMID: 26267322 PMCID: PMC4694906 DOI: 10.18632/oncotarget.4713] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/08/2015] [Indexed: 12/25/2022] Open
Abstract
Ovarian cancer is associated with increased expression of the pro-angiogenic chemokine interleukin-8 (IL-8, CXCL8), which induces tumor cell proliferation, angiogenesis, and metastasis. Even though bortezomib (BZ) has shown remarkable anti-tumor activity in hematological malignancies, it has been less effective in ovarian cancer; however, the mechanisms are not understood. We have recently shown that BZ unexpectedly induces the expression of IL-8 in ovarian cancer cells in vitro, by IκB kinase (IKK)-dependent mechanism. Here, we tested the hypothesis that IKK inhibition reduces the IL-8 production and increases BZ effectiveness in reducing ovarian tumor growth in vivo. Our results demonstrate that the combination of BZ and the IKK inhibitor Bay 117085 significantly reduces the growth of ovarian tumor xenografts in nude mice when compared to either drug alone. Mice treated with the BZ/Bay 117085 combination exhibit smallest tumors, and lowest levels of IL-8. Furthermore, the reduced tumor growth in the combination group is associated with decreased tumor levels of S536P-p65 NFκB and its decreased recruitment to IL-8 promoter in tumor tissues. These data provide the first in vivo evidence that combining BZ with IKK inhibitor is effective, and suggest that using IKK inhibitors may increase BZ effectiveness in ovarian cancer treatment.
Collapse
Affiliation(s)
- Bipradeb Singha
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | | | - Sai Phyo
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Atish Patel
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY 11439, USA
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY 11439, USA
| | - Ivana Vancurova
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| |
Collapse
|
5
|
Shanker A, Pellom ST, Dudimah DF, Thounaojam MC, de Kluyver RL, Brooks AD, Yagita H, McVicar DW, Murphy WJ, Longo DL, Sayers TJ. Bortezomib Improves Adoptive T-cell Therapy by Sensitizing Cancer Cells to FasL Cytotoxicity. Cancer Res 2015; 75:5260-72. [PMID: 26494122 PMCID: PMC4681610 DOI: 10.1158/0008-5472.can-15-0794] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 09/23/2015] [Indexed: 01/15/2023]
Abstract
Cancer immunotherapy shows great promise but many patients fail to show objective responses, including in cancers that can respond well, such as melanoma and renal adenocarcinoma. The proteasome inhibitor bortezomib sensitizes solid tumors to apoptosis in response to TNF-family death ligands. Because T cells provide multiple death ligands at the tumor site, we investigated the effects of bortezomib on T-cell responses in immunotherapy models involving low-avidity antigens. Bortezomib did not affect lymphocyte or tissue-resident CD11c(+)CD8(+) dendritic cell counts in tumor-bearing mice, did not inhibit dendritic cell expression of costimulatory molecules, and did not decrease MHC class I/II-associated antigen presentation to cognate T cells. Rather, bortezomib activated NF-κB p65 in CD8(+) T cells, stabilizing expression of T-cell receptor CD3ζ and IL2 receptor-α, while maintaining IFNγ secretion to improve FasL-mediated tumor lysis. Notably, bortezomib increased tumor cell surface expression of Fas in mice as well as human melanoma tissue from a responsive patient. In renal tumor-bearing immunodeficient Rag2(-/-) mice, bortezomib treatment after adoptive T-cell immunotherapy reduced lung metastases and enhanced host survival. Our findings highlight the potential of proteasome inhibitors to enhance antitumor T-cell function in the context of cancer immunotherapy.
Collapse
Affiliation(s)
- Anil Shanker
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee. Host-Tumor Interactions Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee. School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee.
| | - Samuel T Pellom
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee. School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee. Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, Tennessee
| | - Duafalia F Dudimah
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee
| | - Menaka C Thounaojam
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee
| | - Rachel L de Kluyver
- Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland
| | - Alan D Brooks
- Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland. Basic Sciences Program, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Daniel W McVicar
- Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland
| | - William J Murphy
- Division of Hematology/Oncology, Departments of Dermatology and Internal Medicine, University of California School of Medicine, Davis, California
| | - Dan L Longo
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Thomas J Sayers
- Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland. Basic Sciences Program, Leidos Biomedical Research, Inc., Frederick, Maryland.
| |
Collapse
|
6
|
Pellom ST, Dudimah DF, Thounaojam MC, Sayers TJ, Shanker A. Modulatory effects of bortezomib on host immune cell functions. Immunotherapy 2015; 7:1011-22. [PMID: 26325610 PMCID: PMC4648628 DOI: 10.2217/imt.15.66] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Bortezomib is an inhibitor of the ubiquitin-proteasome proteolytic pathway responsible for intracellular protein turnover. Cellular proteins controlled by this pathway represent a diverse group of potential therapeutic targets, particularly in cancer cells, which exploit this proteasomal pathway to promote their growth and diminish apoptosis. Along with inhibiting the proteasome and thus sensitizing tumor cells to apoptosis, bortezomib may also have multiple effects on the host immune responses. This review summarizes the effects that bortezomib may play on immune cell subsets in various disease states in modifying lymphocyte receptors, ligands, the expression of various cytokines and chemokines and their downstream signaling. We also propose steps that can be taken to refine combinatorial strategies that include bortezomib to improve current immunotherapeutic approaches.
Collapse
Affiliation(s)
- Samuel Troy Pellom
- Department of Biochemistry & Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
- School of Graduate Studies & Research, Meharry Medical College, Nashville, TN 37208, USA
- Department of Microbiology & Immunology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Duafalia Fred Dudimah
- Department of Biochemistry & Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Menaka Chanu Thounaojam
- Department of Biochemistry & Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Thomas Joseph Sayers
- Cancer & Inflammation Program, National Cancer Institute, Frederick, MD 21702, USA
- Basic Sciences Program, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Anil Shanker
- Department of Biochemistry & Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
- School of Graduate Studies & Research, Meharry Medical College, Nashville, TN 37208, USA
- Host–Tumor Interactions Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
7
|
Apetoh L, Smyth MJ, Drake CG, Abastado JP, Apte RN, Ayyoub M, Blay JY, Bonneville M, Butterfield LH, Caignard A, Castelli C, Cavallo F, Celis E, Chen L, Colombo MP, Comin-Anduix B, Coukos G, Dhodapkar MV, Dranoff G, Frazer IH, Fridman WH, Gabrilovich DI, Gilboa E, Gnjatic S, Jäger D, Kalinski P, Kaufman HL, Kiessling R, Kirkwood J, Knuth A, Liblau R, Lotze MT, Lugli E, Marincola F, Melero I, Melief CJ, Mempel TR, Mittendorf EA, Odun K, Overwijk WW, Palucka AK, Parmiani G, Ribas A, Romero P, Schreiber RD, Schuler G, Srivastava PK, Tartour E, Valmori D, van der Burg SH, van der Bruggen P, van den Eynde BJ, Wang E, Zou W, Whiteside TL, Speiser DE, Pardoll DM, Restifo NP, Anderson AC. Consensus nomenclature for CD8 + T cell phenotypes in cancer. Oncoimmunology 2015; 4:e998538. [PMID: 26137416 DOI: 10.1080/2162402x.2014.998538] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 12/10/2014] [Indexed: 10/23/2022] Open
Abstract
Whereas preclinical investigations and clinical studies have established that CD8+ T cells can profoundly affect cancer progression, the underlying mechanisms are still elusive. Challenging the prevalent view that the beneficial effect of CD8+ T cells in cancer is solely attributable to their cytotoxic activity, several reports have indicated that the ability of CD8+ T cells to promote tumor regression is dependent on their cytokine secretion profile and their ability to self-renew. Evidence has also shown that the tumor microenvironment can disarm CD8+ T cell immunity, leading to the emergence of dysfunctional CD8+ T cells. The existence of different types of CD8+ T cells in cancer calls for a more precise definition of the CD8+ T cell immune phenotypes in cancer and the abandonment of the generic terms "pro-tumor" and "antitumor." Based on recent studies investigating the functions of CD8+ T cells in cancer, we here propose some guidelines to precisely define the functional states of CD8+ T cells in cancer.
Collapse
Affiliation(s)
- Lionel Apetoh
- INSERM; UMR 866 , Dijon, France ; Centre Georges François Leclerc , Dijon, France ; Université de Bourgogne , Dijon, France
| | - Mark J Smyth
- QIMR Berghofer Medical Research Institute , Herston, Queensland, Australia
| | - Charles G Drake
- Sidney Kimmel Comprehensive Cancer Center; Johns Hopkins University School of Medicine , Baltimore, MD, USA
| | - Jean-Pierre Abastado
- Institut de Recherches Internationales Servier ; 53, rue Carnot , Suresnes, France
| | - Ron N Apte
- The Shraga Segal Department of Microbiology; Immunology and Genetics ; The Faculty of Health Sciences, Ben Gurion University of the Negev , Beer Sheva, Israel
| | - Maha Ayyoub
- INSERM, Unité1102; Equipe Labellisée Ligue Contre le Cancer ; Institut de Cancérologie de l'Ouest , Nantes-Saint Herblain; France
| | - Jean-Yves Blay
- Cancer Research Center of Lyon; INSERM UMR 1052 ; CNRS UMR 5286 , Centre Leon Berard, Lyon, France ; Medical Oncology Department , Lyon, France
| | - Marc Bonneville
- CRCNA, INSERM U892; CNRS UMR 6299 , Nantes, France ; Institut Mérieux , Lyon, France
| | - Lisa H Butterfield
- University of Pittsburgh Cancer Institute, Departments of Medicine, Surgery, and Immunology , Pittsburgh, PA, USA
| | | | - Chiara Castelli
- Unit of Immunotherapy of Human Tumor; Department of Experimental Oncology and Molecular Medicine ; Fondazione IRCCS Istituto Nazionale dei Tumori , Milan, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences; Molecular Biotechnology Center, University of Torino , Italy
| | - Esteban Celis
- Cancer Immunology; Inflammation and Tolerance Program; Georgia Regents University Cancer Center ; Augusta, GA, USA
| | - Lieping Chen
- Department of Immunobiology and Yale Cancer Center; Yale University School of Medicine , New Haven, CT, USA
| | - Mario P Colombo
- Molecular Immunology Unit; Department of Experimental Oncology and Molecular Medicine ; Fondazione IRCCS Istituto Nazionale dei Tumori ; Milan, Italy
| | - Begoña Comin-Anduix
- UCLA School of Medicine ; Jonsson Comprehensive Cancer Center Los Angeles , CA, USA
| | - Georges Coukos
- Ludwig Center for Cancer Research; Department of Oncology; University of Lausanne , Switzerland
| | - Madhav V Dhodapkar
- Department of Immunobiology and Yale Cancer Center; Yale University School of Medicine , New Haven, CT, USA
| | - Glenn Dranoff
- Department of Medical Oncology and Cancer Vaccine Center; Dana-Farber Cancer Institute and Department of Medicine ; Brigham and Women's Hospital and Harvard Medical School , Boston, MA, USA
| | - Ian H Frazer
- The University of Queensland , Queensland, Australia
| | - Wolf-Hervé Fridman
- Cordeliers Research Centre, University of Paris-Descartes , Paris, France
| | | | - Eli Gilboa
- Department of Microbiology & Immunology; Dodson Interdisciplinary Immunotherapy Institute ; Sylvester Comprehensive Cancer Center; Miller School of Medicine ; University of Miami , Miami, FL, USA
| | - Sacha Gnjatic
- Tisch Cancer Institute; Icahn School of Medicine at Mount Sinai , New York, NY, USA
| | - Dirk Jäger
- Department of Medical Oncology; National Center for Tumor Diseases ; Internal Medicine VI; Heidelberg University Hospital , Heidelberg, Germany
| | - Pawel Kalinski
- Department of Surgery; University of Pittsburgh ; Pittsburgh, PA, USA
| | | | - Rolf Kiessling
- Department of Oncology/Pathology; Karolinska Institutet , Stockholm, Sweden
| | - John Kirkwood
- Division of Hematology/Oncology; Department of Medicine ; School of Medicine; University of Pittsburgh , Pittsburgh; PA; USA ; Melanoma and Skin Cancer Program; University of Pittsburgh Cancer Institute , Pittsburgh, PA, USA
| | | | - Roland Liblau
- INSERM-UMR 1043 ; Toulouse, France ; CNRS ; U5282 , Toulouse, France ; Universite de Toulouse; UPS ; Centre de Physiopathologie Toulouse Purpan (CPTP) ; Toulouse, France ; CHU Toulouse Purpan ; Toulouse, France
| | - Michael T Lotze
- Hillman Cancer Center; University of Pittsburgh Schools of Health Sciences , Pittsburgh, PA, USA
| | - Enrico Lugli
- Unit of Clinical and Experimental Immunology; Humanitas Clinical and Research Center , Rozzano, Italy
| | | | - Ignacio Melero
- Division of Oncology; Center for Applied Medical Research and Clinica Universidad de Navarra , Pamploma, Spain
| | | | - Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases; Massachusetts General Hospital ; Harvard Medical School , Boston, MA, USA
| | - Elizabeth A Mittendorf
- Deparment of Surgical Oncology; University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Kunle Odun
- Departments of Gynecologic Oncology and Immunology; Roswell Park Cancer Institute , Buffalo, NY, USA
| | - Willem W Overwijk
- Department of Melanoma Medical Oncology; University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | | | - Giorgio Parmiani
- Division of Medical Oncology and Immunotherapy; University Hospital , Siena, Italy
| | - Antoni Ribas
- UCLA School of Medicine ; Jonsson Comprehensive Cancer Center Los Angeles , CA, USA
| | - Pedro Romero
- Ludwig Center for Cancer Research; Department of Oncology; University of Lausanne , Switzerland
| | - Robert D Schreiber
- Department of Pathology and Immunology; Washington University School of Medicine , St. Louis, MO USA
| | - Gerold Schuler
- Department of Dermatology; Universitatsklinikum Erlangen , Erlangen, Germany
| | - Pramod K Srivastava
- Center for Immunotherapy of Cancer and Infectious Diseases; Carole and Ray Neag Comprehensive Cancer Center ; University of Connecticut Health Center , Farmington, CT, USA
| | - Eric Tartour
- Department of Clinical Oncology, INSERM U970; Universite Paris Descartes ; Sorbonne Paris-Cité; Paris ; France; Hôpital Européen Georges Pompidou ; Service d'Immunologie Biologique ; Paris, France
| | - Danila Valmori
- INSERM, Unité1102; Equipe Labellisée Ligue Contre le Cancer ; Institut de Cancérologie de l'Ouest , Nantes-Saint Herblain; France ; Faculty of Medicine, University of Nantes, 44035 Nantes, France
| | | | - Pierre van der Bruggen
- Ludwig Institute for Cancer Research; BrusselsBranch de Duve Institute ; Université Catholique de Louvain , Brussels, Blegium
| | - Benoît J van den Eynde
- Ludwig Institute for Cancer Research; BrusselsBranch de Duve Institute ; Université Catholique de Louvain , Brussels, Blegium
| | - Ena Wang
- Research Branch; Sidra Medical and Research Centre , Doha, Qatar
| | - Weiping Zou
- Department of Surgery; University of Michigan School of Medicine , Ann Arbor , MI, USA
| | - Theresa L Whiteside
- Department of Pathology; Immunology, and Otolaryngology ; University of Pittsburgh Cancer Institute , Pittsburgh, PA, USA
| | - Daniel E Speiser
- Ludwig Center for Cancer Research; Department of Oncology; University of Lausanne , Switzerland
| | - Drew M Pardoll
- Sidney Kimmel Comprehensive Cancer Center; Johns Hopkins University School of Medicine , Baltimore, MD, USA
| | - Nicholas P Restifo
- National Cancer Institute; National Institutes of Health , Bethesda, MD, USA
| | - Ana C Anderson
- Evergrande Center for Immunologic Diseases; Ann Romney Center for Neurologic Diseases ; Brigham and Women's Hospital and Harvard Medical School , Boston, MA USA
| |
Collapse
|
8
|
Hersey P, Kakavand H, Wilmott J, van der Westhuizen A, Gallagher S, Gowrishankar K, Scolyer R. How anti-PD1 treatments are changing the management of melanoma. Melanoma Manag 2014; 1:165-172. [PMID: 30190821 PMCID: PMC6094707 DOI: 10.2217/mmt.14.14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The introduction of immunotherapy based on the blockade of the PD1/PD-L1 checkpoints has been associated with high response rates and durable remissions of disease in patients with metastatic melanoma, to the extent that it is now considered the standard of care for a wide range of patients, irrespective of their BRAF or NRAS mutation status. In addition, more frequent follow-up of patients who are at high risk of recurrence after surgical treatment appears to be justified, as does neoadjuvant treatments in order to render patients treatable by surgery. The limitations of this treatment include failure of some patients to respond, a low rate of complete responses and relapses of the disease during treatment. New initiatives in order to overcome these limitations include the identification of biomarkers for the selection responders and evaluations of treatment combinations that will increase responses and their durability. The latter includes combinations with antibodies against other checkpoints on T cells and cotreatments with inhibitors of resistance pathways in melanoma.
Collapse
Affiliation(s)
- Peter Hersey
- Kolling Institute, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
- Melanoma Institute of Australia, Rocklands Road, North Sydney, NSW, Australia
| | - Hojabr Kakavand
- Melanoma Institute of Australia, Rocklands Road, North Sydney, NSW, Australia
- Department of Anatomical Pathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - James Wilmott
- Melanoma Institute of Australia, Rocklands Road, North Sydney, NSW, Australia
- Department of Anatomical Pathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | | | - Stuart Gallagher
- Kolling Institute, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | | | - Richard Scolyer
- Melanoma Institute of Australia, Rocklands Road, North Sydney, NSW, Australia
- Department of Anatomical Pathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| |
Collapse
|
9
|
Škrott Z, Cvek B. Linking the activity of bortezomib in multiple myeloma and autoimmune diseases. Crit Rev Oncol Hematol 2014; 92:61-70. [PMID: 24890785 DOI: 10.1016/j.critrevonc.2014.05.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/25/2014] [Accepted: 05/02/2014] [Indexed: 01/12/2023] Open
Abstract
Since their introduction to the clinic 10 years ago, proteasome inhibitors have become the cornerstone of anti-multiple myeloma therapy. Despite significant progress in understanding the consequences of proteasome inhibition, the unique activity of bortezomib is still unclear. Disappointing results from clinical trials with bortezomib in other malignancies raise the question of what makes multiple myeloma so sensitive to proteasome inhibition. Successful administration of bortezomib in various immunological disorders that exhibit high antibody production suggests that the balance between protein synthesis and degradation is a key determinant of sensitivity to proteasome inhibition because a high rate of protein production is a shared characteristic in plasma and myeloma cells. Initial or acquired resistance to bortezomib remains a major obstacle in the clinic as in vitro data from cell lines suggest a key role for the β5 subunit mutation in resistance; however the mutation was not found in patient samples. Recent studies indicate the importance of selecting for a subpopulation of cells that produce lower amounts of paraprotein during bortezomib therapy.
Collapse
Affiliation(s)
- Zdeněk Škrott
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 11, 78371 Olomouc, Czech Republic
| | - Boris Cvek
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 11, 78371 Olomouc, Czech Republic.
| |
Collapse
|
10
|
Jazirehi AR, Kurdistani SK, Economou JS. Histone deacetylase inhibitor sensitizes apoptosis-resistant melanomas to cytotoxic human T lymphocytes through regulation of TRAIL/DR5 pathway. THE JOURNAL OF IMMUNOLOGY 2014; 192:3981-9. [PMID: 24639349 DOI: 10.4049/jimmunol.1302532] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Modern immune therapies (PD-1/PD-L1 and CTLA-4 checkpoints blockade and adoptive cell transfer) have remarkably improved the response rates of metastatic melanoma. These modalities rely on the killing potential of CTL as proximal mediator of antimelanoma responses. Mechanisms of tumor resistance to and the predominant cytotoxic pathway(s) used by melanoma-reactive CTL are important outcome determinants. We hypothesized that downmodulation of death receptors (DRs) in addition to aberrant apoptotic signaling might confer resistance to death signals delivered by CTL. To test these two hypotheses, we used an in vitro model of MART CTL-resistant melanoma sublines. TCR-transgenic and patient-derived CTLs used the TRAIL cytotoxic pathway through DR5. Furthermore, recombinant human TRAIL and drozitumab (anti-DR5 agonistic mAb) were used to explicitly verify the contribution of the DR5/TRAIL pathway in killing melanomas. CTL resistance was due to DR5 downregulation and an inverted ratio of pro- to antiapoptotic molecules, both of which were reversed by the histone deacetylase inhibitor suberoylanilide hydroxanic acid. Apoptosis negative (c-IAP-2 and Bcl-xL) and positive (DR5) regulators were potential incriminators partly regulating CTL sensitivity. These preclinical findings suggest that exposure to this chromatin remodeling drug of immune-resistant melanomas can skew toward an intracellular proapoptotic milieu, increase DR expression, and overcome acquired immune resistance.
Collapse
Affiliation(s)
- Ali R Jazirehi
- Department of Surgery, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | | | | |
Collapse
|
11
|
Jazirehi AR, Nazarian R, Torres-Collado AX, Economou JS. Aberrant apoptotic machinery confers melanoma dual resistance to BRAF(V600E) inhibitor and immune effector cells: immunosensitization by a histone deacetylase inhibitor. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2014; 3:43-56. [PMID: 24660121 PMCID: PMC3960761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 02/13/2014] [Indexed: 06/03/2023]
Abstract
BRAF(V600E)-inhibitors (BRAFi; e.g., vemurafenib) and modern immune-based therapies such as PD-1/PD-L1 and CTLA-4 checkpoints blockade and adoptive cell transfer (ACT) have significantly improved the care of melanoma patients. Having these two effective (BRAFi and immunotherapy) therapies raises the question whether there is a rational biological basis for using them in combination. We developed an in vitro model to determine whether tumor resistance mechanisms to a small molecule inhibitor of a driver oncogene, and to cytotoxic T lymphocyte (CTL)- and natural killer (NK) cell-delivered apoptotic death signals were exclusive or intersecting. We generated melanoma sublines resistant to BRAFi vemurafenib and to CTL recognizing the MART-1 melanoma antigen. Vemurafenib-resistant (VemR) sublines were cross-resistant to MART CTL and NK cells indicating that a common apoptotic pathway governing tumor response to both modalities was disrupted. Pretreatment of VemR melanomas with a histone deacetylase inhibitor (HDACi) restored sensitivity to MART CTL and NK apoptosis by skewing the apoptotic gene programs towards a proapoptotic phenotype. Our in vitro findings suggest that during the course of acquisition of BRAFi resistance, melanomas develop cross-resistance to CTL- and NK-killing. Further, aberrant apoptotic pathways, amenable by an FDA-approved chromatin remodeling drug, regulate tumor resistance mechanisms to immune effector cells. These results may provide rational molecular basis for further investigations to combine these therapies clinically.
Collapse
Affiliation(s)
- Ali R Jazirehi
- Department of Surgery, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of CaliforniaLos Angeles 90095, USA
| | - Ramin Nazarian
- Department of Medicine, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of CaliforniaLos Angeles 90095, USA
| | - Antoni Xavier Torres-Collado
- Department of Surgery, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of CaliforniaLos Angeles 90095, USA
| | - James S Economou
- Department of Surgery, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of CaliforniaLos Angeles 90095, USA
- Department of Microbiology, Immunology and Molecular Genetics, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of CaliforniaLos Angeles 90095, USA
- Department of Molecular and Medical Pharmacology, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of CaliforniaLos Angeles 90095, USA
| |
Collapse
|
12
|
Shanker A, Dudimah DF, Pellom ST, Uzhachenko RV, Carbone DP, Dikov MM. Cancer therapy by resuscitating Notch immune surveillance. J Immunother Cancer 2014. [PMCID: PMC4081851 DOI: 10.1186/2051-1426-2-s1-o1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
|
13
|
Carozzi VA, Renn CL, Bardini M, Fazio G, Chiorazzi A, Meregalli C, Oggioni N, Shanks K, Quartu M, Serra MP, Sala B, Cavaletti G, Dorsey SG. Bortezomib-induced painful peripheral neuropathy: an electrophysiological, behavioral, morphological and mechanistic study in the mouse. PLoS One 2013; 8:e72995. [PMID: 24069168 PMCID: PMC3772181 DOI: 10.1371/journal.pone.0072995] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 07/23/2013] [Indexed: 11/18/2022] Open
Abstract
Bortezomib is the first proteasome inhibitor with significant antineoplastic activity for the treatment of relapsed/refractory multiple myeloma as well as other hematological and solid neoplasms. Peripheral neurological complications manifesting with paresthesias, burning sensations, dysesthesias, numbness, sensory loss, reduced proprioception and vibratory sensitivity are among the major limiting side effects associated with bortezomib therapy. Although bortezomib-induced painful peripheral neuropathy is clinically easy to diagnose and reliable models are available, its pathophysiology remains partly unclear. In this study we used well-characterized immune-competent and immune-compromised mouse models of bortezomib-induced painful peripheral neuropathy. To characterize the drug-induced pathological changes in the peripheral nervous system, we examined the involvement of spinal cord neuronal function in the development of neuropathic pain and investigated the relevance of the immune response in painful peripheral neuropathy induced by bortezomib. We found that bortezomib treatment induced morphological changes in the spinal cord, dorsal roots, dorsal root ganglia (DRG) and peripheral nerves. Neurophysiological abnormalities and specific functional alterations in Aδ and C fibers were also observed in peripheral nerve fibers. Mice developed mechanical allodynia and functional abnormalities of wide dynamic range neurons in the dorsal horn of spinal cord. Bortezomib induced increased expression of the neuronal stress marker activating transcription factor-3 in most DRG. Moreover, the immunodeficient animals treated with bortezomib developed a painful peripheral neuropathy with the same features observed in the immunocompetent mice. In conclusion, this study extends the knowledge of the sites of damage induced in the nervous system by bortezomib administration. Moreover, a selective functional vulnerability of peripheral nerve fiber subpopulations was found as well as a change in the electrical activity of wide dynamic range neurons of dorsal horn of spinal cord. Finally, the immune response is not a key factor in the development of morphological and functional damage induced by bortezomib in the peripheral nervous system.
Collapse
Affiliation(s)
- Valentina A. Carozzi
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
- * E-mail: (VAC)
| | - Cynthia L. Renn
- School of Nursing, Center for Pain Studies, University of Maryland, Baltimore, Maryland, United States of America
| | - Michela Bardini
- “M. Tettamanti” Research Center, Department of Health Sciences, University of Milan Bicocca, Monza, Italy
| | - Grazia Fazio
- “M. Tettamanti” Research Center, Department of Health Sciences, University of Milan Bicocca, Monza, Italy
| | - Alessia Chiorazzi
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Cristina Meregalli
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Norberto Oggioni
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Kathleen Shanks
- School of Nursing, Center for Pain Studies, University of Maryland, Baltimore, Maryland, United States of America
| | - Marina Quartu
- Department of Biomedical Sciences, Section of Cytomorphology, University of Cagliari, Monserrato, Italy
| | - Maria Pina Serra
- Department of Biomedical Sciences, Section of Cytomorphology, University of Cagliari, Monserrato, Italy
| | - Barbara Sala
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Guido Cavaletti
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Susan G. Dorsey
- School of Nursing, Center for Pain Studies, University of Maryland, Baltimore, Maryland, United States of America
| |
Collapse
|
14
|
Sarhan D, Wennerberg E, D’Arcy P, Gurajada D, Linder S, Lundqvist A. A novel inhibitor of proteasome deubiquitinating activity renders tumor cells sensitive to TRAIL-mediated apoptosis by natural killer cells and T cells. Cancer Immunol Immunother 2013; 62:1359-68. [PMID: 23689729 PMCID: PMC11029014 DOI: 10.1007/s00262-013-1439-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 05/14/2013] [Indexed: 11/30/2022]
Abstract
The proteasome inhibitor bortezomib simultaneously renders tumor cells sensitive to killing by natural killer (NK) cells and resistant to killing by tumor-specific T cells. Here, we show that b-AP15, a novel inhibitor of proteasome deubiquitinating activity, sensitizes tumors to both NK and T cell-mediated killing. Exposure to b-AP15 significantly increased the susceptibility of tumor cell lines of various origins to NK (p < 0.0002) and T cell (p = 0.02)-mediated cytotoxicity. Treatment with b-AP15 resulted in increased tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor-2 expression (p = 0.03) and decreased cFLIP expression in tumor cells in vitro. In tumor-bearing SCID/Beige mice, treatment with b-AP15 followed by infusion of either human NK cells or tumor-specific T cells resulted in a significantly delayed tumor progression compared with mice treated with NK cells (p = 0.006), T cells (p < 0.0001) or b-AP15 alone (p = 0.003). Combined infusion of NK and T cells in tumor-bearing BALB/c mice following treatment with b-AP15 resulted in a significantly prolonged long-term survival compared with mice treated with b-AP15 and NK or T cells (p ≤ 0.01). Our findings show that b-AP15-induced sensitization to TRAIL-mediated apoptosis could be used as a novel strategy to augment the anticancer effects of adoptively infused NK and T cells in patients with cancer.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis/immunology
- Blotting, Western
- Cell Line, Tumor
- Cells, Cultured
- Combined Modality Therapy
- Cytokines/immunology
- Cytokines/metabolism
- Cytotoxicity, Immunologic/drug effects
- Cytotoxicity, Immunologic/immunology
- HCT116 Cells
- HeLa Cells
- Humans
- Immunotherapy, Adoptive
- K562 Cells
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/transplantation
- MCF-7 Cells
- Mice
- Mice, Inbred BALB C
- Mice, SCID
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/therapy
- Piperidones/immunology
- Piperidones/pharmacology
- Protease Inhibitors/immunology
- Protease Inhibitors/pharmacology
- Proteasome Endopeptidase Complex/immunology
- Proteasome Endopeptidase Complex/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/transplantation
- TNF-Related Apoptosis-Inducing Ligand/immunology
- TNF-Related Apoptosis-Inducing Ligand/pharmacology
- Ubiquitination/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Dhifaf Sarhan
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, R8:01, 171 76 Stockholm, Sweden
| | - Erik Wennerberg
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, R8:01, 171 76 Stockholm, Sweden
| | - Padraig D’Arcy
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, R8:01, 171 76 Stockholm, Sweden
| | - Deepthy Gurajada
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, R8:01, 171 76 Stockholm, Sweden
| | - Stig Linder
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, R8:01, 171 76 Stockholm, Sweden
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, R8:01, 171 76 Stockholm, Sweden
| |
Collapse
|