1
|
Moquist PN, Zhang X, Leiske CI, Eng-Duncan NML, Zeng W, Bindman NA, Wo SW, Wong A, Henderson CM, Crowder K, Lyon R, Doronina SO, Senter PD, Neff-LaFord HD, Sussman D, Gardai SJ, Levengood MR. Reversible Chemical Modification of Antibody Effector Function Mitigates Unwanted Systemic Immune Activation. Bioconjug Chem 2024; 35:855-866. [PMID: 38789102 PMCID: PMC11191404 DOI: 10.1021/acs.bioconjchem.4c00212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Antibody effector functions including antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) are mediated through the interaction of the antibody Fc region with Fcγ receptors present on immune cells. Several approaches have been used to modulate antibody Fc-Fcγ interactions with the goal of driving an effective antitumor immune response, including Fc point mutations and glycan modifications. However, robust antibody-Fcγ engagement and immune cell binding of Fc-enhanced antibodies in the periphery can lead to the unwanted induction of systemic cytokine release and other dose-limiting infusion-related reactions. Creating a balance between effective engagement of Fcγ receptors that can induce antitumor activity without incurring systemic immune activation is an ongoing challenge in the field of antibody and immuno-oncology therapeutics. Herein, we describe a method for the reversible chemical modulation of antibody-Fcγ interactions using simple poly(ethylene glycol) (PEG) linkers conjugated to antibody interchain disulfides with maleimide attachments. This method enables dosing of a therapeutic with muted Fcγ engagement that is restored in vivo in a time-dependent manner. The technology was applied to an effector function enhanced agonist CD40 antibody, SEA-CD40, and experiments demonstrate significant reductions in Fc-induced immune activation in vitro and in mice and nonhuman primates despite showing retained efficacy and improved pharmacokinetics compared to the parent antibody. We foresee that this simple, modular system can be rapidly applied to antibodies that suffer from systemic immune activation due to peripheral FcγR binding immediately upon infusion.
Collapse
Affiliation(s)
- Philip N. Moquist
- ADC
Chemistry, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United states
| | - Xinqun Zhang
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Chris I. Leiske
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | | | - Weiping Zeng
- ADC
In Vivo Pharmacology, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Noah A. Bindman
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Serena W. Wo
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Abbie Wong
- ADC
Translational Sciences, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Clark M. Henderson
- ADC
Translational Sciences, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Karalyne Crowder
- Non-Clinical
Sciences, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Robert Lyon
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Svetlana O. Doronina
- ADC
Chemistry, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United states
| | - Peter D. Senter
- ADC
Chemistry, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United states
| | - Haley D. Neff-LaFord
- Non-Clinical
Sciences, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Django Sussman
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Shyra J. Gardai
- Immunology, Pfizer,
Inc., 21823 30th Dr.
SE, Bothell, Washington 98021, United States
| | - Matthew R. Levengood
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| |
Collapse
|
2
|
Barboy O, Bercovich A, Li H, Eyal-Lubling Y, Yalin A, Shapir Itai Y, Abadie K, Zada M, David E, Shlomi-Loubaton S, Katzenelenbogen Y, Jaitin DA, Gur C, Yofe I, Feferman T, Cohen M, Dahan R, Newell EW, Lifshitz A, Tanay A, Amit I. Modeling T cell temporal response to cancer immunotherapy rationalizes development of combinatorial treatment protocols. NATURE CANCER 2024; 5:742-759. [PMID: 38429414 DOI: 10.1038/s43018-024-00734-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 01/19/2024] [Indexed: 03/03/2024]
Abstract
Successful immunotherapy relies on triggering complex responses involving T cell dynamics in tumors and the periphery. Characterizing these responses remains challenging using static human single-cell atlases or mouse models. To address this, we developed a framework for in vivo tracking of tumor-specific CD8+ T cells over time and at single-cell resolution. Our tools facilitate the modeling of gene program dynamics in the tumor microenvironment (TME) and the tumor-draining lymph node (tdLN). Using this approach, we characterize two modes of anti-programmed cell death protein 1 (PD-1) activity, decoupling induced differentiation of tumor-specific activated precursor cells from conventional type 1 dendritic cell (cDC1)-dependent proliferation and recruitment to the TME. We demonstrate that combining anti-PD-1 therapy with anti-4-1BB agonist enhances the recruitment and proliferation of activated precursors, resulting in tumor control. These data suggest that effective response to anti-PD-1 therapy is dependent on sufficient influx of activated precursor CD8+ cells to the TME and highlight the importance of understanding system-level dynamics in optimizing immunotherapies.
Collapse
Affiliation(s)
- Oren Barboy
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Akhiad Bercovich
- Department of Computer Science and Applied Mathematics and Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hanjie Li
- Department of Synthetic Immunology, Shenzhen Institutes of Advanced Technology, Shenzhen, China
| | - Yaniv Eyal-Lubling
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Adam Yalin
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yuval Shapir Itai
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Kathleen Abadie
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Mor Zada
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal David
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shir Shlomi-Loubaton
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Diego Adhemar Jaitin
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Chamutal Gur
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
- The Hebrew University, Jerusalem, Israel
| | - Ido Yofe
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Feferman
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Merav Cohen
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rony Dahan
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Evan W Newell
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, USA
| | - Aviezer Lifshitz
- Department of Computer Science and Applied Mathematics and Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Amos Tanay
- Department of Computer Science and Applied Mathematics and Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
3
|
Naini AA, Mayanti T, Maharani R, Harneti D, Nurlelasari, Farabi K, Fajriah S, Hilmayanti E, Kabayama K, Shimoyama A, Manabe Y, Fukase K, Jungsuttiwong S, Prescott TAK, Supratman U. Paraxylines A-G: Highly oxygenated preurianin-type limonoids with immunomodulatory TLR4 and cytotoxic activities from the stem bark of Dysoxylum parasiticum. PHYTOCHEMISTRY 2024; 220:114009. [PMID: 38342289 DOI: 10.1016/j.phytochem.2024.114009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 02/13/2024]
Abstract
Seven previously undescribed preurianin-type limonoids, namely paraxylines A-G, and three known analogs were isolated from stem bark of Dysoxylum parasiticum. The structures, including absolute configurations, were established through spectroscopic analyses, quantum chemical calculations using the density functional theory method, as well as the DP4+ algorithm. Paraxylines A-G were identified as the first preurianin-type with full substitution at C, D-rings, leading to the highly oxygenated seco-limonoids skeleton. The secreted alkaline phosphate assay against an engineered human and murine TLR4 of HEK-Blue cells was performed to evaluate the immune regulating effects. Among them, paraxyline B was found to be a remarkable TLR4 agonist whereas two analogs (toonapubesins A and B) were found to antagonise lipopolysaccharide stimulation of the TLR4 pathway. Paraxylines A and C-E acted either as agonists or antagonists depending on the origin of the TLR4 receptor (human or mouse). The effect of these selected compounds on the expression of pro-inflammatory cytokines TNF-α, IL-1α, IL-1β, and IL-6 of the NF-κB signaling pathway were examined in macrophage cell lines, revealing dose-dependent effects. Additionally, paraxylines A, C, D, and G also presented modest cytotoxic activity against MCF-7 and HeLa cell lines with IC50 values ranging from 23.1 to 43.5 μM.
Collapse
Affiliation(s)
- Al Arofatus Naini
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia; Central Laboratory, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Tri Mayanti
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia; Study Centre of Natural Product Chemistry and Synthesis, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Rani Maharani
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia; Study Centre of Natural Product Chemistry and Synthesis, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Desi Harneti
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia; Study Centre of Natural Product Chemistry and Synthesis, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Nurlelasari
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia; Study Centre of Natural Product Chemistry and Synthesis, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Kindi Farabi
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia; Study Centre of Natural Product Chemistry and Synthesis, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia
| | - Sofa Fajriah
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Cibinong Science Center Complex - BRIN, Cibinong, 16911, Bogor, West Java, Indonesia
| | - Erina Hilmayanti
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka, 560-0043, Japan
| | - Kazuya Kabayama
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka, 560-0043, Japan
| | - Atsushi Shimoyama
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka, 560-0043, Japan
| | - Yoshiyuki Manabe
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka, 560-0043, Japan
| | - Koichi Fukase
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka, 560-0043, Japan
| | - Sirriporn Jungsuttiwong
- Department of Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani, 34190, Thailand
| | | | - Unang Supratman
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia; Central Laboratory, Universitas Padjadjaran, Jatinangor, 45363, Sumedang, West Java, Indonesia.
| |
Collapse
|
4
|
Zaman R, Islam RA, Chowdhury EH. Evolving therapeutic proteins to precisely kill cancer cells. J Control Release 2022; 351:779-804. [DOI: 10.1016/j.jconrel.2022.09.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 10/31/2022]
|
5
|
Blokon-Kogan D, Levi-Mann M, Malka-Levy L, Itzhaki O, Besser MJ, Shiftan Y, Szöőr Á, Vereb G, Gross G, Abken H, Weinstein-Marom H. Membrane anchored IL-18 linked to constitutively active TLR4 and CD40 improves human T cell antitumor capacities for adoptive cell therapy. J Immunother Cancer 2022. [PMCID: PMC9442493 DOI: 10.1136/jitc-2020-001544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BackgroundAdoptive transfer of tumor-infiltrating lymphocytes (TILs) or blood T cells genetically redirected by an antitumor TCR or CAR induces a strong antitumor response in a proportion of patients with cancer; however, the therapeutic efficacy is often limited by rapid decline in T cell functions. Coadministering supportive cytokines frequently provokes systemic side effects preventing their broad clinical application. We recently showed that cytokines can be anchored to the cell membrane in a functional fashion and that cytokine receptor signaling can synergize with TLR4 and CD40 signaling. Here, we aimed at augmenting T cell activation by simultaneous signaling through the cytokine receptor, toll-like receptor and TNF-type receptor using IL-18, TLR4 and CD40 as prototypes.MethodsGenes were expressed on electroporation of in vitro-transcribed mRNA in CD4+ and CD8+ T cells from healthy donors redirected against melanoma cells with an anti-melanotransferrin CAR and in TILs derived from melanoma patients. Functional assays included the activation of signaling pathways, expression of activation and differentiation markers, cytokine secretion and killing of melanoma target cells.ResultsTo provide IL-18 costimulation to T cells in-cis while avoiding systemic effects, we genetically anchored IL-18 to the T cell membrane, either alone (memIL-18) or fused with constitutively active (ca)TLR4 and caCD40 signaling domains arranged in tandem, creating a synthetic ‘all-in-one’ memIL-18-TLR4-CD40 receptor. MemIL-18-TLR4-CD40, but not memIL-18, triggered strong NF-κB activation in cells lacking the IL-18 receptor, attesting to functionality of the TLR-CD40 moiety. While the membrane-anchored cytokine was found to act mainly in-cis, some T cell activation in-trans was also observed. The electroporated T cells exhibited spontaneous T-bet upregulation and IFN-γ and TNF-α secretion. Melanoma-induced activation of CAR-T cells and TILs as manifested by cytokine secretion and cytolytic activity was substantially augmented by both constructs, with memIL-18-TLR4-CD40 exerting stronger effects than memIL-18 alone.ConclusionsLinking membrane anchored IL-18 with caTLR4 and caCD40 signaling in one hybrid transmembrane protein provides simultaneous activation of three T cell costimulatory pathways through one genetically engineered membrane molecule, strongly amplifying T cell functions for adoptive T cell therapy of cancer.
Collapse
Affiliation(s)
- Dayana Blokon-Kogan
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- The Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee, Israel
| | - Maya Levi-Mann
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- The Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee, Israel
| | - Lior Malka-Levy
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- The Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee, Israel
| | - Orit Itzhaki
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center at Tel Hashomer, Ramat Gan, Israel
| | - Michal J Besser
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center at Tel Hashomer, Ramat Gan, Israel
- Department of Clinical Microbiology and Immunology, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
- Davidoff Center, Rabin Medical Center, Petah Tikvah, Israel
| | - Yuval Shiftan
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
| | - Árpád Szöőr
- Department of Biophysics and Cell Biology, University of Debrecen Faculty of Medicine, Debrecen, Hungary
| | - György Vereb
- Department of Biophysics and Cell Biology, University of Debrecen Faculty of Medicine, Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, University of Debrecen Faculty of Medicine, Debrecen, Hungary
| | - Gideon Gross
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- The Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee, Israel
| | - Hinrich Abken
- Div. Genetic Immunotherapy, Leibniz Institute for Immunotherapy and University Regensburg, Regensburg, Germany
| | - Hadas Weinstein-Marom
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- The Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee, Israel
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center at Tel Hashomer, Ramat Gan, Israel
- Department of Clinical Microbiology and Immunology, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| |
Collapse
|
6
|
A Comparative View on Molecular Alterations and Potential Therapeutic Strategies for Canine Oral Melanoma. Vet Sci 2021; 8:vetsci8110286. [PMID: 34822659 PMCID: PMC8619620 DOI: 10.3390/vetsci8110286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/14/2022] Open
Abstract
Canine oral melanoma (COM) is a highly aggressive tumour associated with poor prognosis due to metastasis and resistance to conventional anti-cancer therapies. As with human mucosal melanoma, the mutational landscape is predominated by copy number aberrations and chromosomal structural variants, but differences in study cohorts and/or tumour heterogeneity can lead to discordant results regarding the nature of specific genes affected. This review discusses somatic molecular alterations in COM that result from single nucleotide variations, copy number changes, chromosomal rearrangements, and/or dysregulation of small non-coding RNAs. A cross-species comparison highlights notable recurrent aberrations, and functionally grouping dysregulated proteins reveals unifying biological pathways that may be critical for oncogenesis and metastasis. Finally, potential therapeutic strategies are considered to target these pathways in canine patients, and the benefits of collaboration between science, medical, and veterinary communities are emphasised.
Collapse
|
7
|
Harnessing the combined potential of cancer immunotherapy and nanomedicine: A new paradigm in cancer treatment. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 40:102492. [PMID: 34775062 DOI: 10.1016/j.nano.2021.102492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 10/16/2021] [Accepted: 10/29/2021] [Indexed: 11/21/2022]
Abstract
Cancer immunotherapy has recently emerged as a rising star due to its ability to activate patients' immune systems to fight tumors and prevent relapse. Conversely, the interest in cancer nanomedicine has seemingly waned due to its lackluster clinical translation. Despite being hailed as a game-changer in oncology, cancer immunotherapy still faces numerous challenges. Combining both entities together has thus been one among several solutions proposed to circumvent these challenges. This solution has since gained traction and has also led to a renaissance of cancer nanomedicine. While most combinations are currently experimental at best, some have progressed on to clinical trials. This review thus seeks to examine the advantages and disadvantages of integrating both modalities as a cancer treatment. The opportunities, challenges and future directions of this emerging field will also be explored with the hope that such a combination will lead to a paradigm shift in cancer treatments.
Collapse
|
8
|
Tang L, Zhang R, Zhang X, Yang L. Personalized Neoantigen-Pulsed DC Vaccines: Advances in Clinical Applications. Front Oncol 2021; 11:701777. [PMID: 34381724 PMCID: PMC8350509 DOI: 10.3389/fonc.2021.701777] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/12/2021] [Indexed: 02/05/2023] Open
Abstract
In the past few decades, great progress has been made in the clinical application of dendritic cell (DC) vaccines loaded with personalized neoantigens. Personalized neoantigens are antigens arising from somatic mutations in cancers, with specificity to each patient. DC vaccines work based on the fundamental characteristics of DCs, which are professional antigen-presenting cells (APCs), responsible for the uptake, processing, and presentation of antigens to T cells to activate immune responses. Neoantigens can exert their antitumor effects only after they are taken up by APCs and presented to T cells. In recent years, neoantigen-based personalized tumor therapeutic vaccines have proven to be safe, immunogenic and feasible treatment strategies in patients with melanoma and glioblastoma that provide new hope in the treatment of cancer patients and a new approach to cure cancer. In addition, according to ClinicalTrials.gov, hundreds of registered DC vaccine trials are either completed or ongoing worldwide, of which 9 are in early phase I, 191 in phase I, 166 in phase II and 8 in phase III. Hundreds of clinical studies on therapeutic tumor vaccines globally have proven that DC vaccines are stable, reliable and very safe. However, in this process, many other factors still limit the effectiveness of the vaccine. This review will focus on the current research progress on personalized neoantigen-pulsed DC vaccines, their limitations and future research directions of DC vaccines loaded with neoantigens. This review aims to provide a better understanding of DCs biology and manipulation of activated DCs for DCs researchers to produce the next generation of highly efficient cancer vaccines for patients.
Collapse
Affiliation(s)
- Lin Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Rui Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xiaoyu Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
9
|
Bonfiglio CA, Weber C, Atzler D, Lutgens E. Immunotherapy and cardiovascular diseases (CVD): novel avenues for immunotherapeutic approaches. QJM 2021; 116:271-278. [PMID: 34293177 DOI: 10.1093/qjmed/hcab207] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/27/2021] [Indexed: 12/20/2022] Open
Abstract
As current therapies for cardiovascular disease (CVD), predominantly based on lipid lowering, still face an unacceptable residual risk, novel treatment strategies are being explored. Besides lipids, inflammatory processes play a major role in the pathogenesis of atherosclerosis, the underlying cause of the majority of CVD. The first clinical trials targeting the interleukin-1β-inflammasome axis have shown that targeting this pathway is successful in reducing cardiovascular events but did not decrease overall CVD mortality. Hence, novel and improved immunotherapeutics to treat CVD are being awaited. In this review we highlight novel immunotherapeutic approaches in CVD as well as future challenges ahead.
Collapse
Affiliation(s)
- Cecilia Assunta Bonfiglio
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Pettenkoferstraße 8a & 9, Munich, 80336, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Pettenkoferstraße 8a & 9, Munich, 80336, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstraße 8a & 9, Munich, 80336, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER, Maastricht University, Maastricht, the Netherlands
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Pettenkoferstraße 8a & 9, Munich, 80336, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstraße 8a & 9, Munich, 80336, Germany
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Goethestraße 33D, Munich, 80336, Germany
| | - Esther Lutgens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Pettenkoferstraße 8a & 9, Munich, 80336, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstraße 8a & 9, Munich, 80336, Germany
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, AZ Amsterdam, 1105, The Netherlands
| |
Collapse
|
10
|
Kimura T, Goi T, Yokoi S, Ohnishi K, Togawa T, Iida A, Ishida M, Sato Y. Possible spontaneous regression of hepatocellular carcinoma with unique histopathological features confirmed by surgical resection: a case report. Surg Case Rep 2021; 7:162. [PMID: 34255193 PMCID: PMC8276907 DOI: 10.1186/s40792-021-01246-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/04/2021] [Indexed: 11/12/2022] Open
Abstract
Background Spontaneous regression of hepatocellular carcinoma (HCC) is a rare event, and its clinicopathological features and underlying mechanism are not fully understood. Case presentation An 84-year-old female with hepatitis C virus infection and diabetes mellitus was referred to our hospital for further examination. Abdominal ultrasonography showed a 3.4-cm solid tumor with a heterogeneous irregular center and no fibrous capsule in liver segment 8 (S8). An enhanced computed tomography (CT) scan revealed a tumor in S8 with heterogeneous enhancement in the arterial phase and washed out insufficiently in the portal and equilibrium phase. The enhanced pattern on magnetic resonance imaging was similar to that of CT. Although the imaging findings were not typical for HCC, liver resection (S8) was performed with HCC as the most probable diagnosis. Histopathological examination of the resected specimen showed that the tumor was well to moderately differentiated HCC with unique features. Approximately half of the tumor was composed of well-differentiated HCC that was focally accompanied by dense lymphocyte infiltration. The other half of the tumor was fibrotic tissue that resembled an inflammatory pseudotumor. Several foci of moderately differentiated HCC were scattered within the tumor with a nodule-in-nodule appearance, and the foci totally showed coagulative necrosis. On immunostaining, lymphocytes in the tumor stroma were positive for CD8 and programmed death 1. The expression of programmed death-ligand 1 was observed in carcinoma cells and macrophages specifically within the lymphocyte-rich area of HCC. Conclusions We consider this case representative of spontaneous regression of HCC, and the immune response against HCC might contribute to tumor regression, leading to complex histopathological appearances. This case may provide insight into the mechanism of spontaneous regression of HCC.
Collapse
Affiliation(s)
- Toshihisa Kimura
- Department of Surgery, National Hospital Organization, Tsuruga Medical Center, 33-1, Sakuragaoka, Tsuruga, Fukui, 914-0195, Japan. .,First Department of Surgery, Faculty of Medicine, University of Fukui, 23-3, Matsuoka Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan.
| | - Takanori Goi
- First Department of Surgery, Faculty of Medicine, University of Fukui, 23-3, Matsuoka Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Shigehiro Yokoi
- First Department of Surgery, Faculty of Medicine, University of Fukui, 23-3, Matsuoka Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Kenji Ohnishi
- Department of Surgery, National Hospital Organization, Tsuruga Medical Center, 33-1, Sakuragaoka, Tsuruga, Fukui, 914-0195, Japan
| | - Tamotsu Togawa
- Department of Surgery, National Hospital Organization, Tsuruga Medical Center, 33-1, Sakuragaoka, Tsuruga, Fukui, 914-0195, Japan
| | - Atsushi Iida
- Department of Surgery, National Hospital Organization, Tsuruga Medical Center, 33-1, Sakuragaoka, Tsuruga, Fukui, 914-0195, Japan
| | - Makoto Ishida
- Department of Surgery, Tannan Regional Medical Center, 1-2-31, Saburoku-cho, Sabae, Fukui, 916-8515, Japan
| | - Yasunori Sato
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, 13-1, Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| |
Collapse
|
11
|
Manjunath M, Choudhary B. Triple-negative breast cancer: A run-through of features, classification and current therapies. Oncol Lett 2021; 22:512. [PMID: 33986872 PMCID: PMC8114477 DOI: 10.3892/ol.2021.12773] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 03/01/2021] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the most prevalent cancer in women worldwide. Triple-negative breast cancer (TNBC) is characterized by the lack of expression of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2. It is the most aggressive subtype of breast cancer and accounts for 12-20% of all breast cancer cases. TNBC is associated with younger age of onset, greater metastatic potential, higher incidence of relapse, and lower overall survival rates. Based on molecular phenotype, TNBC has been classified into six subtypes (BL1, BL2, M, MES, LAR, and IM). TNBC treatment is challenging due to its heterogeneity, highly invasive nature, and relatively poor therapeutics response. Chemotherapy and radiotherapy are conventional strategies for the treatment of TNBC. Recent research in TNBC and mechanistic understanding of disease pathogenesis using cutting-edge technologies has led to the unfolding of new lines of therapies that have been incorporated into clinical practice. Poly (ADP-ribose) polymerase and immune checkpoint inhibitors have made their way to the current TNBC treatment paradigm. This review focuses on the classification, features, and treatment progress in TNBC. Histological subtypes connected to recurrence, molecular classification of TNBC, targeted therapy for early and advanced TNBC, and advances in non-coding RNA in therapy are the key highlights in this review.
Collapse
Affiliation(s)
- Meghana Manjunath
- Department of Biotechnology, Institute of Bioinformatics and Applied Biotechnology, Bengaluru, Karnataka 560100, India
- Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Bibha Choudhary
- Department of Biotechnology, Institute of Bioinformatics and Applied Biotechnology, Bengaluru, Karnataka 560100, India
| |
Collapse
|
12
|
Chauhan DS, Dhasmana A, Laskar P, Prasad R, Jain NK, Srivastava R, Jaggi M, Chauhan SC, Yallapu MM. Nanotechnology synergized immunoengineering for cancer. Eur J Pharm Biopharm 2021; 163:72-101. [PMID: 33774162 PMCID: PMC8170847 DOI: 10.1016/j.ejpb.2021.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/06/2021] [Accepted: 03/15/2021] [Indexed: 12/26/2022]
Abstract
Novel strategies modulating the immune system yielded enhanced anticancer responses and improved cancer survival. Nevertheless, the success rate of immunotherapy in cancer treatment has been below expectation(s) due to unpredictable efficacy and off-target effects from systemic dosing of immunotherapeutic(s). As a result, there is an unmet clinical need for improving conventional immunotherapy. Nanotechnology offers several new strategies, multimodality, and multiplex biological targeting advantage to overcome many of these challenges. These efforts enable programming the pharmacodynamics, pharmacokinetics, and delivery of immunomodulatory agents/co-delivery of compounds to prime at the tumor sites for improved therapeutic benefits. This review provides an overview of the design and clinical principles of biomaterials driven nanotechnology and their potential use in personalized nanomedicines, vaccines, localized tumor modulation, and delivery strategies for cancer immunotherapy. In this review, we also summarize the latest highlights and recent advances in combinatorial therapies availed in the treatment of cold and complicated tumors. It also presents key steps and parameters implemented for clinical success. Finally, we analyse, discuss, and provide clinical perspectives on the integrated opportunities of nanotechnology and immunology to achieve synergistic and durable responses in cancer treatment.
Collapse
Affiliation(s)
- Deepak S Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Partha Laskar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Rajendra Prasad
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Nishant K Jain
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Murali M Yallapu
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA.
| |
Collapse
|
13
|
Rückert M, Flohr AS, Hecht M, Gaipl US. Radiotherapy and the immune system: More than just immune suppression. STEM CELLS (DAYTON, OHIO) 2021; 39:1155-1165. [PMID: 33961721 DOI: 10.1002/stem.3391] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/20/2021] [Indexed: 11/07/2022]
Abstract
Radiotherapy (RT) is still one of the standard cancer therapies, with up to two third of all cancer patients with solid tumors being irradiated in the course of their disease. The aim of using ionizing radiation in fractionated treatment schedules was always to achieve local tumor control by inducing DNA damage which can be repaired by surrounding normal tissue but leads to cell death in tumor cells. Meanwhile, it is known that RT also has immunological effects reshaping the tumor microenvironment. Nevertheless, RT alone often fails to elicit potent antitumor immune responses as these effects can be immunostimulatory as well as immunosuppressive. Here, we discuss how immunotherapies can be exploited in combined therapies to boost RT-induced antitumor immune responses or to counteract preexisting and RT-mediated immunosuppression to improve local and systemic tumor control. Furthermore, we highlight some parameters of radioimmunotherapies (RITs) which are under investigation for potential optimizations and how RIT approaches are tested in first phases II and III trials. Finally, we discuss how RT might affect normal and cancer stem cells.
Collapse
Affiliation(s)
- Michael Rückert
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Ann-Sophie Flohr
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Markus Hecht
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Udo S Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Erlangen, Germany
| |
Collapse
|
14
|
Guo D, Ji X, Luo J. Rational nanocarrier design towards clinical translation of cancer nanotherapy. Biomed Mater 2021; 16. [DOI: 10.1088/1748-605x/abe35a] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/04/2021] [Indexed: 02/06/2023]
|
15
|
Wang T, Xu X, Zhang K. Nanotechnology-Enabled Chemodynamic & Immunotherapy. Curr Cancer Drug Targets 2021; 21:545-557. [PMID: 33618647 DOI: 10.2174/1568009621666210219101552] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/07/2022]
Abstract
High-level reactive oxygen species (ROS) have been reported to exert a robust anti-tumor effect by inducing cell apoptosis or necroptosis. Based on the Fenton reaction or Fenton-like reaction, a therapeutic strategy (i.e., chemodynamic therapy (CDT)) is proposed, where hydroxyl radicals (•OH) that are one typical ROS via the spontaneous activation by endogenous stimulus can be produced to kill tumors. Moreover, high-level ROS can also facilitate tumor-associated antigen exposure, which benefits phagocytosis of corpses and debris by antigen-presenting cells (e.g., dendritic cells (DCs)) and further activates systematic immune responses. Great efforts wherein nanotechnology is underlined have been made in interdisciplinary communities to witness the development of this field. To provide a comprehensive understanding of CDT, the state of art of strategies on nanotechnology-enabled CDT is discussed in detail. In particular, the combination of CDT and its augmented immunotherapy against tumor for overcoming the poor outcome that mono-CDT suffers from is highlighted. Moreover, the potential challenges will also be discussed.
Collapse
Affiliation(s)
- Taixia Wang
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Tongji University School of Medicine, 301 Yan-chang-zhong Road, Shanghai, 200072. China
| | - Xiaohong Xu
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Tongji University School of Medicine, 301 Yan-chang-zhong Road, Shanghai, 200072. China
| | - Kun Zhang
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Tongji University School of Medicine, 301 Yan-chang-zhong Road, Shanghai, 200072. China
| |
Collapse
|
16
|
Choudhry H. The Microbiome and Its Implications in Cancer Immunotherapy. Molecules 2021; 26:E206. [PMID: 33401586 PMCID: PMC7795182 DOI: 10.3390/molecules26010206] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/22/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is responsible for ~18 million deaths globally each year, representing a major cause of death. Several types of therapy strategies such as radiotherapy, chemotherapy and more recently immunotherapy, have been implemented in treating various types of cancer. Microbes have recently been found to be both directly and indirectly involved in cancer progression and regulation, and studies have provided novel and clear insights into the microbiome-mediated emergence of cancers. Scientists around the globe are striving hard to identify and characterize these microbes and the underlying mechanisms by which they promote or suppress various kinds of cancer. Microbes may influence immunotherapy by blocking various cell cycle checkpoints and the production of certain metabolites. Hence, there is an urgent need to better understand the role of these microbes in the promotion and suppression of cancer. The identification of microbes may help in the development of future diagnostic tools to cure cancers possibly associated with the microbiome. This review mainly focuses on various microbes and their association with different types of cancer, responses to immunotherapeutic modulation, physiological responses, and prebiotic and postbiotic effects.
Collapse
Affiliation(s)
- Hani Choudhry
- Department of Biochemistry, Faculty of Sciences, Cancer and Mutagenesis Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
17
|
Abstract
Modern cancer immunotherapy has revolutionised oncology and carries the potential to radically change the approach to cancer treatment. However, numerous questions remain to be answered to understand immunotherapy response better and further improve the benefit for future cancer patients. Computational models are promising tools that can contribute to accelerated immunotherapy research by providing new clues and hypotheses that could be tested in future trials, based on preceding simulations in addition to the empirical rationale. In this topical review, we briefly summarise the history of cancer immunotherapy, including computational modelling of traditional cancer immunotherapy, and comprehensively review computational models of modern cancer immunotherapy, such as immune checkpoint inhibitors (as monotherapy and combination treatment), co-stimulatory agonistic antibodies, bispecific antibodies, and chimeric antigen receptor T cells. The modelling approaches are classified into one of the following categories: data-driven top-down vs mechanistic bottom-up, simplistic vs detailed, continuous vs discrete, and hybrid. Several common modelling approaches are summarised, such as pharmacokinetic/pharmacodynamic models, Lotka-Volterra models, evolutionary game theory models, quantitative systems pharmacology models, spatio-temporal models, agent-based models, and logic-based models. Pros and cons of each modelling approach are critically discussed, particularly with the focus on the potential for successful translation into immuno-oncology research and routine clinical practice. Specific attention is paid to calibration and validation of each model, which is a necessary prerequisite for any successful model, and at the same time, one of the main obstacles. Lastly, we provide guidelines and suggestions for the future development of the field.
Collapse
Affiliation(s)
- Damijan Valentinuzzi
- Jožef Stefan Institute, Jamova cesta 39, 1000 Ljubljana, Slovenia. Faculty of Mathematics and Physics, University of Ljubljana, Jadranska ulica 19, 1111 Ljubljana, Slovenia
| | | |
Collapse
|
18
|
Bonfá G, Blazquez-Roman J, Tarnai R, Siciliano V. Precision Tools in Immuno-Oncology: Synthetic Gene Circuits for Cancer Immunotherapy. Vaccines (Basel) 2020; 8:E732. [PMID: 33287392 PMCID: PMC7761833 DOI: 10.3390/vaccines8040732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 12/16/2022] Open
Abstract
Engineered mammalian cells for medical purposes are becoming a clinically relevant reality thanks to advances in synthetic biology that allow enhanced reliability and safety of cell-based therapies. However, their application is still hampered by challenges including time-consuming design-and-test cycle iterations and costs. For example, in the field of cancer immunotherapy, CAR-T cells targeting CD19 have already been clinically approved to treat several types of leukemia, but their use in the context of solid tumors is still quite inefficient, with additional issues related to the adequate quality control for clinical use. These limitations can be overtaken by innovative bioengineering approaches currently in development. Here we present an overview of recent synthetic biology strategies for mammalian cell therapies, with a special focus on the genetic engineering improvements on CAR-T cells, discussing scenarios for the next generation of genetic circuits for cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Velia Siciliano
- Synthetic and Systems Biology Lab for Biomedicine, Istituto Italiano di Tecnologia-IIT, Largo Barsanti e Matteucci, 80125 Naples, Italy; (G.B.); (J.B.-R.); (R.T.)
| |
Collapse
|
19
|
McAvoy MB, Choi BD, Jones PS. Immune Therapy for Central Nervous System Metastasis. Neurosurg Clin N Am 2020; 31:627-639. [PMID: 32921357 DOI: 10.1016/j.nec.2020.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Brain metastases lead to substantial morbidity and mortality among patients with advanced malignancies. Although treatment options have traditionally included largely palliative measures, studies of brain metastasis response to immunotherapy are promising. Immune checkpoint inhibitors have shown efficacy in studies of patients with melanoma, renal cell carcinoma, and lung cancer brain metastases. Patients with brain metastases are more frequently included in clinical trials, ushering in a new era in immunotherapy and management for patients with brain metastases. Gaining an understanding of the molecular determination for response to immunotherapies remains a major challenge and is an active area of future research.
Collapse
Affiliation(s)
- Malia B McAvoy
- University of Washington Medical Center, Department of Neurological Surgery, Box 356470, 1959 NE Pacific Street, Seattle, WA 98195-6470, USA
| | - Bryan D Choi
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, 15 Parkman Street, WAC 3, Boston, MA 02114, USA
| | - Pamela S Jones
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, 15 Parkman Street, WAC 745, Boston, MA 02114, USA.
| |
Collapse
|
20
|
Wu Z, Liu J, Dai R, Wu S. Current status and future perspectives of immunotherapy in bladder cancer treatment. SCIENCE CHINA-LIFE SCIENCES 2020; 64:512-533. [PMID: 32926318 DOI: 10.1007/s11427-020-1768-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022]
Abstract
The treatment strategy of bladder cancer has evolved not only through the traditional modalities of surgery and chemotherapy but also by immunotherapy over the past several decades. Immunotherapies such as intravesical Bacillus Calmette-Guérin (BCG) vaccines and immune checkpoint blockades (ICBs) are sometimes used for treating patients with bladder cancer, especially those who develop resistance to conventional first-line treatments such as surgery and chemotherapy. Unfortunately, it is a limited number of individuals that see clinical benefits from this approach, and complicating matters more is that many of these patients suffer severe immune-related adverse events (irAEs). If current momentum continues to result in improved response rates and managed irAEs, immunotherapy could be poised to revolutionize the landscape of urothelial carcinoma therapeutics.
Collapse
Affiliation(s)
- Zhangsong Wu
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China.,Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China
| | - Jinjian Liu
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China.,Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China
| | - Ruixiang Dai
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China.,Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China
| | - Song Wu
- Department of Urological Surgery, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China. .,Shenzhen Following Precision Medical Institute, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China. .,Department of Urological Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, China.
| |
Collapse
|
21
|
Combined Expression of Genetic Adjuvants Via mRNA Electroporation Exerts Multiple Immunostimulatory Effects on Antitumor T Cells. J Immunother 2020; 42:43-50. [PMID: 30489430 DOI: 10.1097/cji.0000000000000252] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Adoptive transfer of tumor-infiltrating lymphocytes (TILs) or gene-modified T cells expressing antitumor TCRs or chimeric antigen receptors often yields a high rate of clinical response in several types of cancer. New approaches for enhancing the functional properties of antitumor T cells could improve the clinical outcome of these treatments. To this end, we created 3 classes of genes, each designed to operate autonomously upon expression in T cells. We recently reported on the enhancing effects of constitutively active toll-like receptor 4 (caTLR4), membrane (mem) interleukin-2, memIL-12, and memIL-15, and self-oligomerizing, constitutively active CD40 (caCD40). Here, we evaluated their combined effects on peripheral blood CD8 T cells and different antimelanoma TIL cultures following mRNA electroporation. Expression in CD8 T cells induced transient production of interferon-γ and prolonged and robust upregulation of CD25, CD69, 4-1BB, and OX40. The adjuvants enhanced cytolytic activity of TILs and production of interferon-γ and TNF-α in the presence of autologous, but not mismatched, melanoma for at least 3 days after electroporation. Expression of the 3 adjuvants in young TILs from different patients markedly increased the expression of CD25, OX40, 4-1BB, CD127, and CD28 and exhibited cooperative and, at times, synergistic effects. Furthermore, predefined mixtures of mRNA encoding these adjuvants markedly enhanced the specific antitumor response of selected TILs and killing of autologous melanoma cells by young TILs. Our findings suggest that combinations of these new genetic adjuvants can substantially improve the functional properties of antitumor T cells, offering a new tool of unique versatility in adoptive cell therapy.
Collapse
|
22
|
Sharma P, Diergaarde B, Ferrone S, Kirkwood JM, Whiteside TL. Melanoma cell-derived exosomes in plasma of melanoma patients suppress functions of immune effector cells. Sci Rep 2020; 10:92. [PMID: 31919420 PMCID: PMC6952363 DOI: 10.1038/s41598-019-56542-4] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/12/2019] [Indexed: 12/21/2022] Open
Abstract
Melanoma patients' plasma contains exosomes produced by malignant and normal cells. Plasma exosomes were isolated and separated by immunocapture into two fractions: melanoma cell-derived exosomes (MTEX) and normal cell-derived exosomes (non-MTEX). Immunosuppressive effects of MTEX on primary human immune cells were evaluated. Exosomes were isolated from plasma of 12 melanoma patients and six healthy donors (HDs). Expression levels of 19 immunoregulatory proteins in MTEX, non-MTEX and HDs exosomes were evaluated by on-bead flow cytometry. Functional/phenotypic changes induced in CD8+ T or natural killer (NK) cells by MTEX or non-MTEX were compared. Plasma protein levels were higher in patients than HDs (P < 0.0009). In patients, MTEX accounted for 23-66% of total exosomes. MTEX were enriched in immunosuppressive proteins (P = 0.03). MTEX, but not HDs exosomes, inhibited CD69 expression (P ≤ 0.0008), induced apoptosis (P ≤ 0.0009) and suppressed proliferation (P ≤ 0.002) in CD8+ T cells and downregulated NKG2D expression in NK cells (P = 0.001). Non-MTEX were enriched in immunostimulatory proteins (P = 0.002) and were only weakly immunosuppressive. Elevated MTEX/total exosome ratios and, surprisingly, non-MTEX ability to induce apoptosis of CD8+ T cells emerged as positive correlates of disease stage. MTEX emerge as the major mechanism of tumor-induced immune suppression and as an underestimated barrier to successful melanoma immunotherapy.
Collapse
Affiliation(s)
- Priyanka Sharma
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Brenda Diergaarde
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh and UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - John M Kirkwood
- Department of Medicine, University of Pittsburgh School of Medicine and UPMC Hillman Cancer, Pittsburgh, PA, 15213, USA
| | - Theresa L Whiteside
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA.
- Departments of Immunology and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
23
|
Genetically Modified T-Cell Therapy for Osteosarcoma: Into the Roaring 2020s. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1257:109-131. [PMID: 32483735 DOI: 10.1007/978-3-030-43032-0_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T-cell immunotherapy may offer an approach to improve outcomes for patients with osteosarcoma who fail current therapies. In addition, it has the potential to reduce treatment-related complications for all patients. Generating tumor-specific T cells with conventional antigen-presenting cells ex vivo is time-consuming and often results in T-cell products with a low frequency of tumor-specific T cells. Furthermore, the generated T cells remain sensitive to the immunosuppressive tumor microenvironment. Genetic modification of T cells is one strategy to overcome these limitations. For example, T cells can be genetically modified to render them antigen specific, resistant to inhibitory factors, or increase their ability to home to tumor sites. Most genetic modification strategies have only been evaluated in preclinical models; however, early clinical phase trials are in progress. In this chapter, we will review the current status of gene-modified T-cell therapy with special focus on osteosarcoma, highlighting potential antigenic targets, preclinical and clinical studies, and strategies to improve current T-cell therapy approaches.
Collapse
|
24
|
Zhao Z, Zheng L, Chen W, Weng W, Song J, Ji J. Delivery strategies of cancer immunotherapy: recent advances and future perspectives. J Hematol Oncol 2019; 12:126. [PMID: 31779642 PMCID: PMC6883629 DOI: 10.1186/s13045-019-0817-3] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/31/2019] [Indexed: 12/25/2022] Open
Abstract
Immunotherapy has become an emerging strategy for the treatment of cancer. Immunotherapeutic drugs have been increasing for clinical treatment. Despite significant advances in immunotherapy, the clinical application of immunotherapy for cancer patients has some challenges associated with safety and efficacy, including autoimmune reactions, cytokine release syndrome, and vascular leak syndrome. Novel strategies, particularly improved delivery strategies, including nanoparticles, scaffolds, and hydrogels, are able to effectively target tumors and/or immune cells of interest, increase the accumulation of immunotherapies within the lesion, and reduce off-target effects. Here, we briefly describe five major types of cancer immunotherapy, including their clinical status, strengths, and weaknesses. Then, we introduce novel delivery strategies, such as nanoparticle-based delivery of immunotherapy, implantable scaffolds, injectable biomaterials for immunotherapy, and matrix-binding molecular conjugates, which can improve the efficacy and safety of immunotherapies. Also, the limitations of novel delivery strategies and challenges of clinical translation are discussed.
Collapse
Affiliation(s)
- Zhongwei Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Liyun Zheng
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Weiqian Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Wei Weng
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Jingjing Song
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China. .,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China. .,Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Affiliated Lishui Hospital of Zhejiang University, The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.
| |
Collapse
|
25
|
Ascione A, Arenaccio C, Mallano A, Flego M, Gellini M, Andreotti M, Fenwick C, Pantaleo G, Vella S, Federico M. Development of a novel human phage display-derived anti-LAG3 scFv antibody targeting CD8 + T lymphocyte exhaustion. BMC Biotechnol 2019; 19:67. [PMID: 31623599 PMCID: PMC6798348 DOI: 10.1186/s12896-019-0559-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 09/09/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Lymphocyte-activation gene (LAG)3 is a 498 aa transmembrane type I protein acting as an immune inhibitory receptor. It is expressed on activated lymphocytes, natural killer cells and plasmacytoid dendritic cells. In activated lymphocytes, LAG3 expression is involved in negative control of cell activation/proliferation to ensure modulation and control of immune responses. In view of its deregulated expression in tumor-infiltrating lymphocytes, LAG3, together with the additional immune checkpoint inhibitors CTLA4 and PD1, is considered a major target in order to reverse the immunosuppression typically mounting in oncologic diseases. Since many patients still fail to respond to current immune checkpoints-based therapies, the identification of new effective immune inhibitors is a priority in the ongoing fight against cancer. RESULTS We identified a novel human single-chain variable fragment (scFv) Ab against a conformational epitope of LAG3 by in vitro phage display technology using the recombinant antigen as a bait. This scFv (referred to as F7) was characterized in terms of binding specificity to both recombinant antigen and human LAG3-expressing cells. It was then rebuilt into an IgG format pre-optimized for clinical usage, and the resulting bivalent construct was shown to preserve its ability to bind LAG3 on human cells. Next, we analyzed the activity of the anti-LAG3 scFvF7 using two different antigen-specific CD8+ T lymphocyte clones as target cells. We proved that the reconstituted anti-LAG3 F7 Ab efficiently binds the cell membrane of both cell clones after peptide-activation. Still more significantly, we observed a striking increase in the peptide-dependent cell activation upon Ab treatment as measured in terms of IFN-γ release by both ELISA and ELISPOT assays. CONCLUSIONS Overall, the biotechnological strategy described herein represents a guiding development model for the search of novel useful immune checkpoint inhibitors. In addition, our functional data propose a novel candidate reagent for consideration as a cancer treatment.
Collapse
Affiliation(s)
- Alessandro Ascione
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy.
| | - Claudia Arenaccio
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Alessandra Mallano
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Michela Flego
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Mara Gellini
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Mauro Andreotti
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Craig Fenwick
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Stefano Vella
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Maurizio Federico
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| |
Collapse
|
26
|
Passariello M, D'Alise AM, Esposito A, Vetrei C, Froechlich G, Scarselli E, Nicosia A, De Lorenzo C. Novel Human Anti-PD-L1 mAbs Inhibit Immune-Independent Tumor Cell Growth and PD-L1 Associated Intracellular Signalling. Sci Rep 2019; 9:13125. [PMID: 31511565 PMCID: PMC6739323 DOI: 10.1038/s41598-019-49485-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/23/2019] [Indexed: 02/08/2023] Open
Abstract
The novel antibody-based immunotherapy in oncology exploits the activation of immune system mediated by immunomodulatory antibodies specific for immune checkpoints. Among them, the programmed death ligand-1 (PD-L1) is of particular interest as it is expressed not only on T-cells, but also on other immune cells and on a large variety of cancer cells, such as breast cancer cells, considering its high expression in both ErbB2-positive and Triple Negative Breast Cancers. We demonstrate here that PD-L1_1, a novel anti-PD-L1 T -cell stimulating antibody, inhibits PD-L1-tumor cell growth also by affecting the intracellular MAPK pathway and by activating caspase 3. Similar in vitro results were obtained for the first time here also with the clinically validated anti-PD-L1 mAb Atezolizumab and in vivo with another validated anti-mouse anti-PD-L1 mAb. Moreover, we found that two high affinity variants of PD-L1_1 inhibited tumor cell viability more efficiently than the parental PD-L1_1 by affecting the same MAPK pathways with a more potent effect. Altogether, these results shed light on the role of PD-L1 in cancer cells and suggest that PD-L1_1 and its high affinity variants could become powerful antitumor weapons to be used alone or in combination with other drugs such as the anti-ErbB2 cAb already successfully tested in in vitro combinatorial treatments.
Collapse
Affiliation(s)
- Margherita Passariello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy.,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy
| | | | - Annachiara Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy.,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Cinzia Vetrei
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy.,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Guendalina Froechlich
- Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy.,European School of Molecular Medicine, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | | | - Alfredo Nicosia
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy.,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy.,Keires AG Bäumleingasse 18, CH-4051, Basel, Switzerland
| | - Claudia De Lorenzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy. .,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy.
| |
Collapse
|
27
|
Passariello M, Camorani S, Vetrei C, Cerchia L, De Lorenzo C. Novel Human Bispecific Aptamer-Antibody Conjugates for Efficient Cancer Cell Killing. Cancers (Basel) 2019; 11:E1268. [PMID: 31470510 PMCID: PMC6770524 DOI: 10.3390/cancers11091268] [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: 06/26/2019] [Revised: 08/19/2019] [Accepted: 08/23/2019] [Indexed: 12/26/2022] Open
Abstract
Monoclonal antibodies have been approved by the Food and Drug Administration for the treatment of various human cancers. More recently, oligonucleotide aptamers have risen increasing attention for cancer therapy thanks to their low size (efficient tumor penetration) and lack of immunogenicity, even though the short half-life and lack of effector functions still hinder their clinical applications. Here, we demonstrate, for the first time, that two novel bispecific conjugates, consisting of an anti-epidermal growth factor receptor (EGFR) aptamer linked either with an anti-epidermal growth factor receptor 2 (ErbB2) compact antibody or with an immunomodulatory (anti-PD-L1) antibody, were easily and rapidly obtained. These novel aptamer-antibody conjugates retain the targeting ability of both the parental moieties and acquire a more potent cancer cell killing activity by combining their inhibitory properties. Furthermore, the conjugation of the anti-EGFR aptamer with the immunomodulatory antibody allowed for the efficient redirection and activation of T cells against cancer cells, thus dramatically enhancing the cytotoxicity of the two conjugated partners. We think that these bispecific antibody-aptamer conjugates could have optimal biological features for therapeutic applications, such as increased specificity for tumor cells expressing both targets and improved pharmacokinetic and pharmacodynamic properties due to the combined advantages of the aptamer and antibody.
Collapse
Affiliation(s)
- Margherita Passariello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131 Napoli, Italy
- Ceinge-Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Simona Camorani
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), CNR, Via S. Pansini 5, 80131 Napoli, Italy
| | - Cinzia Vetrei
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131 Napoli, Italy
- Ceinge-Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Laura Cerchia
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), CNR, Via S. Pansini 5, 80131 Napoli, Italy.
| | - Claudia De Lorenzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131 Napoli, Italy.
- Ceinge-Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145 Naples, Italy.
| |
Collapse
|
28
|
Abstract
Immunotherapy has become a powerful clinical strategy for treating cancer. The number of immunotherapy drug approvals has been increasing, with numerous treatments in clinical and preclinical development. However, a key challenge in the broad implementation of immunotherapies for cancer remains the controlled modulation of the immune system, as these therapeutics have serious adverse effects including autoimmunity and nonspecific inflammation. Understanding how to increase the response rates to various classes of immunotherapy is key to improving efficacy and controlling these adverse effects. Advanced biomaterials and drug delivery systems, such as nanoparticles and the use of T cells to deliver therapies, could effectively harness immunotherapies and improve their potency while reducing toxic side effects. Here, we discuss these research advances, as well as the opportunities and challenges for integrating delivery technologies into cancer immunotherapy, and we critically analyse the outlook for these emerging areas.
Collapse
Affiliation(s)
- Rachel S Riley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert Langer
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
29
|
Mirza AH, Thomas G, Ottensmeier CH, King EV. Importance of the immune system in head and neck cancer. Head Neck 2019; 41:2789-2800. [DOI: 10.1002/hed.25716] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 10/21/2018] [Accepted: 02/07/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
- Adal H. Mirza
- Somers Cancer Research Building MP824; Southampton General Hospital; Southampton United Kingdom
- Department of Head and Neck Surgery; Poole Hospital NHS Foundation Trust; Poole United Kingdom
| | - Gareth Thomas
- Somers Cancer Research Building MP824; Southampton General Hospital; Southampton United Kingdom
| | | | - Emma V. King
- Somers Cancer Research Building MP824; Southampton General Hospital; Southampton United Kingdom
- Department of Head and Neck Surgery; Poole Hospital NHS Foundation Trust; Poole United Kingdom
| |
Collapse
|
30
|
Combining radiation therapy and cancer immune therapies: From preclinical findings to clinical applications. Cancer Radiother 2018; 22:567-580. [PMID: 30197026 DOI: 10.1016/j.canrad.2018.07.136] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 07/23/2018] [Indexed: 12/17/2022]
Abstract
Besides its direct cytotoxic effect on the tumor cells, radiation therapy is also able to elicit an immune-mediated systemic anti-tumor response, resulting in tumor regression in irradiated sites but also within distant out of field secondary lesions and providing a long-term anti-tumor response. It is now clear that associating ionizing radiation with immune therapies can enhance radio-induced anti-tumor immune responses. Over the last decade, such a combination aroused considerable interest among the scientific community, with many preclinical models and clinical trials, using many types of immune therapies and radiation regimens. In this article, we summarize the main mechanisms underlying radio-induced anti-tumor responses. We will then present an extended overview of the recent preclinical and clinical research built on this background of combined radiation and immune therapy, shedding light on what we know so far about such a promising strategy.
Collapse
|
31
|
Sasso E, D'Avino C, Passariello M, D'Alise AM, Siciliano D, Esposito ML, Froechlich G, Cortese R, Scarselli E, Zambrano N, Nicosia A, De Lorenzo C. Massive parallel screening of phage libraries for the generation of repertoires of human immunomodulatory monoclonal antibodies. MAbs 2018; 10:1060-1072. [PMID: 29995563 PMCID: PMC6204801 DOI: 10.1080/19420862.2018.1496772] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoints are emerging as novel targets for cancer therapy, and antibodies against them have shown remarkable clinical efficacy with potential for combination treatments to achieve high therapeutic index. This work aims at providing a novel approach for the generation of several novel human immunomodulatory antibodies capable of binding their targets in their native conformation and useful for therapeutic applications. We performed a massive parallel screening of phage libraries by using for the first time activated human lymphocytes to generate large collections of single-chain variable fragments (scFvs) against 10 different immune checkpoints: LAG-3, PD-L1, PD-1, TIM3, BTLA, TIGIT, OX40, 4-1BB, CD27 and ICOS. By next-generation sequencing and bioinformatics analysis we ranked individual scFvs in each collection and identified those with the highest level of enrichment. As a proof of concept of the quality/potency of the binders identified by this approach, human IgGs from three of these collections (i.e., PD-1, PD-L1 and LAG-3) were generated and shown to have comparable or better binding affinity and biological activity than the clinically validated anti-PD-1 mAb nivolumab. The repertoires generated in this work represent a convenient source of agonistic or antagonistic antibodies against the ‘Checkpoint Immunome’ for preclinical screening and clinical implementation of optimized treatments.
Collapse
Affiliation(s)
- Emanuele Sasso
- a Department of Molecular Medicine and Medical Biotechnology , University of Naples "Federico II" , Napoli ( NA ), Italy.,b CEINGE - Biotecnologie Avanzate s.c. a.r.l ., Naples , Italy
| | - Chiara D'Avino
- a Department of Molecular Medicine and Medical Biotechnology , University of Naples "Federico II" , Napoli ( NA ), Italy.,b CEINGE - Biotecnologie Avanzate s.c. a.r.l ., Naples , Italy
| | - Margherita Passariello
- a Department of Molecular Medicine and Medical Biotechnology , University of Naples "Federico II" , Napoli ( NA ), Italy.,b CEINGE - Biotecnologie Avanzate s.c. a.r.l ., Naples , Italy
| | | | - Daniela Siciliano
- a Department of Molecular Medicine and Medical Biotechnology , University of Naples "Federico II" , Napoli ( NA ), Italy.,b CEINGE - Biotecnologie Avanzate s.c. a.r.l ., Naples , Italy
| | | | - Guendalina Froechlich
- a Department of Molecular Medicine and Medical Biotechnology , University of Naples "Federico II" , Napoli ( NA ), Italy.,b CEINGE - Biotecnologie Avanzate s.c. a.r.l ., Naples , Italy
| | | | | | - Nicola Zambrano
- a Department of Molecular Medicine and Medical Biotechnology , University of Naples "Federico II" , Napoli ( NA ), Italy.,b CEINGE - Biotecnologie Avanzate s.c. a.r.l ., Naples , Italy
| | - Alfredo Nicosia
- a Department of Molecular Medicine and Medical Biotechnology , University of Naples "Federico II" , Napoli ( NA ), Italy.,b CEINGE - Biotecnologie Avanzate s.c. a.r.l ., Naples , Italy.,e Keires AG , Basel , Switzerland
| | - Claudia De Lorenzo
- a Department of Molecular Medicine and Medical Biotechnology , University of Naples "Federico II" , Napoli ( NA ), Italy.,b CEINGE - Biotecnologie Avanzate s.c. a.r.l ., Naples , Italy
| |
Collapse
|
32
|
Dorta-Estremera S, Chin RL, Sierra G, Nicholas C, Yanamandra AV, Nookala SMK, Yang G, Singh S, Curran MA, Sastry KJ. Mucosal HPV E6/E7 Peptide Vaccination in Combination with Immune Checkpoint Modulation Induces Regression of HPV + Oral Cancers. Cancer Res 2018; 78:5327-5339. [PMID: 30054333 DOI: 10.1158/0008-5472.can-18-0892] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/15/2018] [Accepted: 07/17/2018] [Indexed: 12/22/2022]
Abstract
High-risk human papillomavirus (HPV)-associated squamous cell carcinomas of the oropharynx (SCCOP) are among the fastest growing cancers. After standard-of-care treatment, however, patients with HPV+ SCCOP have better overall and disease-specific survival than patients with HPV- SCCOP, suggesting the importance of HPV-specific immunity. We reasoned that therapeutic vaccination targeting the HPV-16 E6 and E7 oncogenes could elicit high-affinity, high-frequency tumor antigen-specific T-cell responses, which could then be augmented and shielded from suppression in the tumor microenvironment by immune checkpoint modulation. In this study, we used a preclinical syngeneic mouse model of oral cancer comprised of mouse tonsil-derived epithelial cells stably expressing HPV-16 E6 and E7 genes along with H-ras oncogene (mEER) to identify combinations of vaccination and checkpoint antibodies capable of promoting tumor regression. Intranasal HPV E6/E7 peptide vaccination and single checkpoint antibodies failed to elicit responses in more than half of animals; however, 4-1BB agonist antibody along with either CD40 agonist antibody or CTLA-4 blockade eliminated the majority of established mEER tumors. The combination of intranasal HPV peptide vaccine and α4-1BB and αCTLA-4 antibodies produced curative efficacy and a better safety profile against orally implanted mEER tumors. Correlates of protective immunity included enhanced intratumoral levels of CD8 T cells relative to immunosuppressive regulatory T cells and myeloid-derived suppressor cells. Overall, our results demonstrate combination vaccine-immunotherapy modalities as novel treatment options for HPV+ SCCOP.Significance: Combinations of vaccine and checkpoint modulation are effective and safe treatment options for HPV+ oral cancers. Cancer Res; 78(18); 5327-39. ©2018 AACR.
Collapse
Affiliation(s)
| | - Renee L Chin
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gloria Sierra
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Courtney Nicholas
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ananta V Yanamandra
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sita M K Nookala
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Guojun Yang
- The University of Texas MD Anderson Cancer Center, Oncology Research for Biologics and Immunotherapy Translation, Houston, Texas, Texas
| | - Shail Singh
- Department of Melanoma and Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, Texas
| | - Michael A Curran
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - K Jagannadha Sastry
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences at Houston, Houston, Texas
| |
Collapse
|
33
|
Bordry N, Broggi MAS, de Jonge K, Schaeuble K, Gannon PO, Foukas PG, Danenberg E, Romano E, Baumgaertner P, Fankhauser M, Wald N, Cagnon L, Abed-Maillard S, Maby-El Hajjami H, Murray T, Ioannidou K, Letovanec I, Yan P, Michielin O, Matter M, Swartz MA, Speiser DE. Lymphatic vessel density is associated with CD8 + T cell infiltration and immunosuppressive factors in human melanoma. Oncoimmunology 2018; 7:e1462878. [PMID: 30221058 PMCID: PMC6136869 DOI: 10.1080/2162402x.2018.1462878] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/25/2018] [Accepted: 03/13/2018] [Indexed: 12/11/2022] Open
Abstract
Increased density of tumor-associated lymphatic vessels correlates with poor patient survival in melanoma and other cancers, yet lymphatic drainage is essential for initiating an immune response. Here we asked whether and how lymphatic vessel density (LVD) correlates with immune cell infiltration in primary tumors and lymph nodes (LNs) from patients with cutaneous melanoma. Using immunohistochemistry and quantitative image analysis, we found significant positive correlations between LVD and CD8+ T cell infiltration as well as expression of the immunosuppressive molecules inducible nitric oxide synthase (iNOS) and 2,3-dioxygénase (IDO). Interestingly, similar associations were seen in tumor-free LNs adjacent to metastatic ones, indicating loco-regional effects of tumors. Our data suggest that lymphatic vessels play multiple roles at tumor sites and LNs, promoting both T cell infiltration and adaptive immunosuppressive mechanisms. Lymph vessel associated T cell infiltration may increase immunotherapy success rates provided that the treatment overcomes adaptive immune resistance.
Collapse
Affiliation(s)
- Natacha Bordry
- Clinical Tumor Biology and Immunotherapy Group, Department of Oncology and Ludwig Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
- Institute of Bioengineering and Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Maria A. S. Broggi
- Institute of Bioengineering and Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Kaat de Jonge
- Clinical Tumor Biology and Immunotherapy Group, Department of Oncology and Ludwig Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Karin Schaeuble
- Clinical Tumor Biology and Immunotherapy Group, Department of Oncology and Ludwig Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Philippe O. Gannon
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Periklis G. Foukas
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
- Department of Surgery, CHUV, Lausanne, Switzerland
| | - Esther Danenberg
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Emanuela Romano
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
- Department of Oncology, INSERM U932, Institut Curie, Paris, FRANCE
| | - Petra Baumgaertner
- Clinical Tumor Biology and Immunotherapy Group, Department of Oncology and Ludwig Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Manuel Fankhauser
- Institute of Bioengineering and Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Noémie Wald
- Clinical Tumor Biology and Immunotherapy Group, Department of Oncology and Ludwig Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Laurène Cagnon
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Samia Abed-Maillard
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Hélène Maby-El Hajjami
- Clinical Tumor Biology and Immunotherapy Group, Department of Oncology and Ludwig Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Timothy Murray
- Clinical Tumor Biology and Immunotherapy Group, Department of Oncology and Ludwig Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Kalliopi Ioannidou
- Clinical Tumor Biology and Immunotherapy Group, Department of Oncology and Ludwig Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | | | - Pu Yan
- Department of Pathology, CHUV, Lausanne, Switzerland
| | - Olivier Michielin
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Maurice Matter
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
- Department of Surgery, CHUV, Lausanne, Switzerland
| | - Melody A. Swartz
- Institute of Bioengineering and Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- 2nd Department of Pathology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Daniel E. Speiser
- Clinical Tumor Biology and Immunotherapy Group, Department of Oncology and Ludwig Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
34
|
High cytotoxic T-lymphocyte-associated antigen 4 and phospho-Akt expression in tumor samples predicts poor clinical outcomes in ipilimumab-treated melanoma patients. Melanoma Res 2018; 27:24-31. [PMID: 27768639 DOI: 10.1097/cmr.0000000000000305] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Ipilimumab, a fully human monoclonal antibody against cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), is the first immune checkpoint inhibitor approved for the treatment of unresectable melanoma on the basis of its overall survival (OS) benefit. However, ipilimumab is associated with significant immune-related adverse events. We hypothesized that biomarker exploration of pretreatment tumor samples and correlation with clinical outcome would enable patient selection with an increased benefit/risk ratio for ipilimumab therapy. At the University of Texas MD Anderson Cancer Center, a total of 81 advanced melanoma patients were treated on the Ipilimumab Expanded Access Program from 2007 to 2008. Using immunohistochemistry, we analyzed the expression of immune checkpoint (CTLA-4, PD-1, PD-L1) and Akt-pathway proteins in formalin-fixed tumor tissue. Associations between these biomarkers and progression-free survival (PFS) and OS were analyzed with univariate and multivariate Cox proportional-hazards models. There was a significant correlation between high CTLA-4 protein expression levels in tumor cells and risk of death (P=0.02) and decreased PFS (P=0.023). In addition, high expression of CTLA-4 in peritumoral lymphocytes correlated with poor OS (P=0.023). In multivariate analysis, patients with high CTLA-4 and phospho-Akt (p-Akt) expression correlated with poor OS (log-rank test, P=0.039) and PFS (log-rank test, P=0.014). High levels of CTLA-4 and p-Akt expression in pretreatment tumor cells in melanoma patients were associated with poor clinical outcomes. Immunohistochemistry analysis of CTLA-4 and p-Akt in pretreatment tumor samples provides useful biomarkers that may enable improved patient selection for ipilimumab therapy. Prospective clinical studies are warranted to investigate the predictive value of these biomarkers.
Collapse
|
35
|
Abstract
Cancer immunotherapy can successfully promote long-term anticancer immune responses, although there is still only a limited number of patients who benefit from such treatment, and it can sometimes have severe treatment-associated adverse events. Compared with systemic immunomodulation, local immunomodulation may enable more effective treatment at lower doses and, at the same time, prevent systemic toxicity. Local delivery of engineered three-dimensional scaffolds may fulfil this role by acting as synthetic immune niches that boost anticancer immunity. In this Opinion article, we highlight the potential of scaffold-based adoptive cell transfer and scaffold-based cancer vaccines that, although applied locally, can promote systemic antitumour immunity. Furthermore, we discuss how scaffold-based cancer immunotherapy may contribute to the development of the next generation of cancer treatments.
Collapse
|
36
|
Yang W, Lu YP, Yang YZ, Kang JR, Jin YD, Wang HW. Expressions of programmed death (PD)-1 and PD-1 ligand (PD-L1) in cervical intraepithelial neoplasia and cervical squamous cell carcinomas are of prognostic value and associated with human papillomavirus status. J Obstet Gynaecol Res 2017; 43:1602-1612. [PMID: 28833798 DOI: 10.1111/jog.13411] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 04/23/2017] [Accepted: 05/03/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Wen Yang
- Department of Gynaecology and Obstetrics; General Hospital of People's Liberation Army; Beijing China
| | - Yan-Ping Lu
- Department of Gynaecology and Obstetrics; General Hospital of People's Liberation Army; Beijing China
| | - Yi-Zhou Yang
- Department of Gynaecology and Obstetrics; General Hospital of People's Liberation Army; Beijing China
| | - Jia-Rui Kang
- Department of Pathology; First Affiliated Hospital of General Hospital of People's Liberation Army; Beijing China
| | - Yi-Duo Jin
- Department of Pathology; First Affiliated Hospital of General Hospital of People's Liberation Army; Beijing China
| | - Hong-Wei Wang
- Department of Pathology; First Affiliated Hospital of General Hospital of People's Liberation Army; Beijing China
| |
Collapse
|
37
|
Xia AL, Wang XC, Lu YJ, Lu XJ, Sun B. Chimeric-antigen receptor T (CAR-T) cell therapy for solid tumors: challenges and opportunities. Oncotarget 2017; 8:90521-90531. [PMID: 29163850 PMCID: PMC5685771 DOI: 10.18632/oncotarget.19361] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/18/2017] [Indexed: 12/11/2022] Open
Abstract
Chimeric antigen receptor (CAR)-engineered T cells (CAR-T cells) have been shown to have unprecedented efficacy in B cell malignancies, most notably in B cell acute lymphoblastic leukemia (B-ALL) with up to a 90% complete remission rate using anti-CD19 CAR-T cells. However, CAR T-cell therapy for solid tumors currently is faced with numerous challenges such as physical barriers, the immunosuppressive tumor microenvironment and the specificity and safety. The clinical results in solid tumors have been much less encouraging, with multiple cases of toxicity and a lack of therapeutic response. In this review, we will discuss the current stats and challenges of CAR-T cell therapy for solid tumors, and propose possibl e solutions and future perspectives.
Collapse
Affiliation(s)
- An-Liang Xia
- Liver Transplantation Center of the First Affiliated Hospital and Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province 210029, P.R. China
| | - Xiao-Chen Wang
- Liver Transplantation Center of the First Affiliated Hospital and Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province 210029, P.R. China
| | - Yi-Jun Lu
- Liver Transplantation Center of the First Affiliated Hospital and Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province 210029, P.R. China
| | - Xiao-Jie Lu
- Liver Transplantation Center of the First Affiliated Hospital and Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province 210029, P.R. China
| | - Beicheng Sun
- Liver Transplantation Center of the First Affiliated Hospital and Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province 210029, P.R. China
| |
Collapse
|
38
|
Immunotherapy targeting immune check-point(s) in brain metastases. Cytokine Growth Factor Rev 2017; 36:33-38. [PMID: 28736183 DOI: 10.1016/j.cytogfr.2017.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 11/21/2022]
Abstract
Immunotherapy with monoclonal antibodies (mAb) directed to different immune check-point(s) is showing a significant clinical impact in a growing number of human tumors of different histotype, both in terms of disease response and long-term survival patients. In this rapidly changing scenario, treatment of brain metastases remains an high unmeet medical need, and the efficacy of immunotherapy in these highly dismal clinical setting remains to be largely demonstrated. Nevertheless, up-coming observations are beginning to suggest a clinical potential of cancer immunotherapy also in brain metastases, regardless the underlying tumor histotype. These observations remain to be validated in larger clinical trials eventually designed also to address the efficacy of therapeutic mAb to immune check-point(s) within multimodality therapies for brain metastases. Noteworthy, the initial proofs of efficacy on immunotherapy in central nervous system metastases are already fostering clinical trials investigating its therapeutic potential also in primary brain tumors. We here review ongoing immunotherapeutic approaches to brain metastases and primary brain tumors, and the foreseeable strategies to overcome their main biologic hurdles and clinical challenges.
Collapse
|
39
|
Terranova-Barberio M, Thomas S, Munster PN. Epigenetic modifiers in immunotherapy: a focus on checkpoint inhibitors. Immunotherapy 2017; 8:705-19. [PMID: 27197539 DOI: 10.2217/imt-2016-0014] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Immune surveillance should be directed to suppress tumor development and progression, involving a balance of coinhibitory and costimulatory signals that amplify immune response without overwhelming the host. Immunotherapy confers durable clinical benefit in 'immunogenic tumors', whereas in other tumors the responses are modest. Thus, immune checkpoint inhibitors may need to be combined with strategies to boost immune response or increase the tumor immune profile. Epigenetic aberrations contribute significantly to carcinogenesis. Recent findings suggest that epigenetic drugs prime the immune response by increasing expression of tumor-associated antigens and immune-related genes, as well as modulating chemokines and cytokines involved in immune system activation. This review describes our current understanding regarding epigenetic and immunotherapy combination, focusing on immune response priming to checkpoint blockade.
Collapse
Affiliation(s)
- Manuela Terranova-Barberio
- Department of Medicine, Division of Hematology & Oncology, University of California, Room A722, 1600 Divisadero St, Box 1770, San Francisco, CA 94115, USA
| | - Scott Thomas
- Department of Medicine, Division of Hematology & Oncology, University of California, Room A722, 1600 Divisadero St, Box 1770, San Francisco, CA 94115, USA
| | - Pamela N Munster
- Department of Medicine, Division of Hematology & Oncology, University of California, Room A722, 1600 Divisadero St, Box 1770, San Francisco, CA 94115, USA
| |
Collapse
|
40
|
Intratumoral modulation of the inducible co-stimulator ICOS by recombinant oncolytic virus promotes systemic anti-tumour immunity. Nat Commun 2017; 8:14340. [PMID: 28194010 PMCID: PMC5316835 DOI: 10.1038/ncomms14340] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 12/19/2016] [Indexed: 12/26/2022] Open
Abstract
Emerging data suggest that locoregional cancer therapeutic approaches with oncolytic viruses can lead to systemic anti-tumour immunity, although the appropriate targets for intratumoral immunomodulation using this strategy are not known. Here we find that intratumoral therapy with Newcastle disease virus (NDV), in addition to the activation of innate immunity, upregulates the expression of T-cell co-stimulatory receptors, with the inducible co-stimulator (ICOS) being most notable. To explore ICOS as a direct target in the tumour, we engineered a recombinant NDV-expressing ICOS ligand (NDV-ICOSL). In the bilateral flank tumour models, intratumoral administration of NDV-ICOSL results in enhanced infiltration with activated T cells in both virus-injected and distant tumours, and leads to effective rejection of both tumours when used in combination with systemic CTLA-4 blockade. These findings highlight that intratumoral immunomodulation with an oncolytic virus expressing a rationally selected ligand can be an effective strategy to drive systemic efficacy of immune checkpoint blockade. Oncolytic viruses induce a variety of immune targets in the infected tumours. Here, the authors show that Newcastle Disease Virus (NDV) upregulates the inducible co-stimulator (ICOS) on T cells and that intratumoral targeting of ICOS with engineered NDV in combination with CTLA-4 blockade induces systemic anti-tumour immunity in mice.
Collapse
|
41
|
Cancer-immune therapy: restoration of immune response in cancer by immune cell modulation. THE NUCLEUS 2017. [DOI: 10.1007/s13237-017-0194-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
42
|
Garnelo M, Tan A, Her Z, Yeong J, Lim CJ, Chen J, Lim KH, Weber A, Chow P, Chung A, Ooi LLPJ, Toh HC, Heikenwalder M, Ng IOL, Nardin A, Chen Q, Abastado JP, Chew V. Interaction between tumour-infiltrating B cells and T cells controls the progression of hepatocellular carcinoma. Gut 2017; 66:342-351. [PMID: 26669617 PMCID: PMC5284473 DOI: 10.1136/gutjnl-2015-310814] [Citation(s) in RCA: 325] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 10/12/2015] [Accepted: 10/14/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The nature of the tumour-infiltrating leucocytes (TILs) is known to impact clinical outcome in carcinomas, including hepatocellular carcinoma (HCC). However, the role of tumour-infiltrating B cells (TIBs) remains controversial. Here, we investigate the impact of TIBs and their interaction with T cells on HCC patient prognosis. DESIGN Tissue samples were obtained from 112 patients with HCC from Singapore, Hong Kong and Zurich and analysed using immunohistochemistry and immunofluorescence. RNA expression of CD19, CD8A, IFNG was analysed using quantitative PCR. The phenotype of freshly isolated TILs was analysed using flow cytometry. A mouse model depleted of mature B cells was used for functional study. RESULTS Tumour-infiltrating T cells and B cells were observed in close contact with each other and their densities are correlated with superior survival in patients with HCC. Furthermore, the density of TIBs was correlated with an enhanced expression of granzyme B and IFN-γ, as well as with reduced tumour viability defined by low expression of Ki-67, and an enhanced expression of activated caspase-3 on tumour cells. CD27 and CD40 costimulatory molecules and TILs expressing activation marker CD38 in the tumour were also correlated with patient survival. Mice depleted of mature B cells and transplanted with murine hepatoma cells showed reduced tumour control and decreased local T cell activation, further indicating the important role of B cells. CONCLUSIONS The close proximity of tumour-infiltrating T cells and B cells indicates a functional interaction between them that is linked to an enhanced local immune activation and contributes to better prognosis for patients with HCC.
Collapse
Affiliation(s)
- Marta Garnelo
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Alex Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Zhisheng Her
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Biopolis, Singapore
| | - Joe Yeong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore,Department of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Chun Jye Lim
- SingHealth Translational Immunology and Inflammation Centre (STIIC), Singapore Health Services Pte Ltd, Singapore, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Kiat Hon Lim
- Department of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Achim Weber
- Institute of Surgical Pathology, University Hospital of Zurich, Zurich, Switzerland
| | - Pierce Chow
- National Cancer Centre, Singapore, Singapore,Singapore General Hospital, Singapore, Singapore,Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Alexander Chung
- National Cancer Centre, Singapore, Singapore,Singapore General Hospital, Singapore, Singapore
| | - London Lucien PJ Ooi
- National Cancer Centre, Singapore, Singapore,Singapore General Hospital, Singapore, Singapore,Duke-NUS Graduate Medical School, Singapore, Singapore
| | | | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany,Institute of Virology, Technical University München/Helmholtz Zentrum München, Germany
| | - Irene O L Ng
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Alessandra Nardin
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Biopolis, Singapore,National Cancer Centre, Singapore, Singapore
| | - Jean-Pierre Abastado
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore,Institut de Recherches Internationales Servier, Suresnes, France
| | - Valerie Chew
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore,SingHealth Translational Immunology and Inflammation Centre (STIIC), Singapore Health Services Pte Ltd, Singapore, Singapore,Duke-NUS Graduate Medical School, Singapore, Singapore
| |
Collapse
|
43
|
Mohammed S, Sukumaran S, Bajgain P, Watanabe N, Heslop HE, Rooney CM, Brenner MK, Fisher WE, Leen AM, Vera JF. Improving Chimeric Antigen Receptor-Modified T Cell Function by Reversing the Immunosuppressive Tumor Microenvironment of Pancreatic Cancer. Mol Ther 2017; 25:249-258. [PMID: 28129119 DOI: 10.1016/j.ymthe.2016.10.016] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 10/07/2016] [Accepted: 10/09/2016] [Indexed: 12/21/2022] Open
Abstract
The adoptive transfer of T cells redirected to tumor-associated antigens via transgenic expression of chimeric antigen receptors (CARs) has produced tumor responses, even in patients with refractory diseases. To target pancreatic cancer, we generated CAR T cells directed against prostate stem cell antigen (PSCA) and demonstrated specific tumor lysis. However, pancreatic tumors employ immune evasion strategies such as the production of inhibitory cytokines, which limit CAR T cell persistence and function. Thus, to protect our cells from the immunosuppressive cytokine IL-4, we generated an inverted cytokine receptor in which the IL-4 receptor exodomain was fused to the IL-7 receptor endodomain (4/7 ICR). Transgenic expression of this molecule in CAR-PSCA T cells should invert the inhibitory effects of tumor-derived IL-4 and instead promote T cell proliferation. We now demonstrate the suppressed activity of CAR T cells in tumor-milieu conditions and the ability of CAR/ICR T cells to thrive in an IL-4-rich microenvironment, resulting in enhanced antitumor activity. Importantly, CAR/ICR T cells remained both antigen and cytokine dependent. These findings support the benefit of combining the 4/7 ICR with CAR-PSCA to treat pancreatic cancer, a PSCA-expressing tumor characterized by a dense immunosuppressive environment rich in IL-4.
Collapse
Affiliation(s)
- Somala Mohammed
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA; Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sujita Sukumaran
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA; Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pradip Bajgain
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA; Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Norihiro Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - William E Fisher
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ann M Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Juan F Vera
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA.
| |
Collapse
|
44
|
Horenstein AL, Chillemi A, Quarona V, Zito A, Mariani V, Faini AC, Morandi F, Schiavoni I, Ausiello CM, Malavasi F. Antibody mimicry, receptors and clinical applications. Hum Antibodies 2017; 25:75-85. [PMID: 28035914 DOI: 10.3233/hab-160305] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
This review focuses on the concept of antibodies acting as receptor agonists and antagonists, and on the potential relevance of this notion in applied medicine. Antibodies are composed of three functional units: two antigen-binding fragments (Fabs) that confer antigen specificity and one constant fragment (Fc) linking antibodies to immune effector functions. The proof-of-concept that large amounts of highly specific and homogeneous antibodies could be produced was provided in 1975 by César Milstein and Georges Köhler. These monoclonal antibody (mAb) reagents started a revolution in medical research, diagnostics, and clinical applications. Alongside diagnostic applications, mAbs were successfully used in vivo: (i) to bind (neutralize/antagonize) antigens expressed on the surface of tumor cells; (ii) to activate immune effector mechanisms; (iii) to crosslink plasma membrane receptors and hence activate therapeutic signaling pathways; and lastly, (iv) the technique was expanded to produce bispecific mAbs, which can bind two different antigens while retaining the ability to activate immune effector functions. The abilities of mAbs to bind, transduce signals, and exert immunostimulatory agonistic capacities are the central issues of this review. The starting point is that some mAbs operate as molecular agonists, substituting for the natural ligand of the receptor. Our analysis is restricted to mAbs that act as receptor agonist/antagonists by either mimicking ligand binding, or through allosteric modulation mediated by binding sites that are topographically distinct from the orthosteric binding site. Functional considerations based on the agonistic stimulation of human CD38 by specific mAbs as surrogate ligands are described as examples of the features of such molecules.
Collapse
Affiliation(s)
- Alberto L Horenstein
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
- CeRMS, University of Torino, Torino 10126, Italy
| | - Antonella Chillemi
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
- CeRMS, University of Torino, Torino 10126, Italy
| | - Valeria Quarona
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
- CeRMS, University of Torino, Torino 10126, Italy
| | - Andrea Zito
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
- CeRMS, University of Torino, Torino 10126, Italy
| | - Valentina Mariani
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
- CeRMS, University of Torino, Torino 10126, Italy
| | - Angelo C Faini
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
- CeRMS, University of Torino, Torino 10126, Italy
| | - Fabio Morandi
- Laboratory of Oncology, Istituto Giannina Gaslini, Genova 16148, Italy
| | - Ilaria Schiavoni
- Department of Infectious, Parasitic, and Immune-Mediated Diseases, Istituto Superiore di Sanità, Roma 00161, Italy
| | - Clara Maria Ausiello
- Department of Infectious, Parasitic, and Immune-Mediated Diseases, Istituto Superiore di Sanità, Roma 00161, Italy
| | - Fabio Malavasi
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Torino 10126, Italy
- CeRMS, University of Torino, Torino 10126, Italy
- Transplantation Immunology, Città della Salute e della Scienza, Torino 10126, Italy
| |
Collapse
|
45
|
Carlo MI, Voss MH, Motzer RJ. Checkpoint inhibitors and other novel immunotherapies for advanced renal cell carcinoma. Nat Rev Urol 2016; 13:420-31. [PMID: 27324121 PMCID: PMC5532875 DOI: 10.1038/nrurol.2016.103] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The management of advanced renal cell carcinoma (RCC) has dramatically changed over the past decade. Therapies that target the vascular endothelial growth factor (VEGF) and mammalian target of rapamycin (mTOR) pathways have considerably expanded treatment options; however, most patients with advanced RCC still have limited overall survival. Increased understanding of the mechanisms of T cell-antigen recognition and function has led to the development of novel immunotherapies to treat cancer, chief among them inhibitors of checkpoint receptors - molecules whose function is to restrain the host immune response. In 2015, the FDA approved the first checkpoint inhibitor nivolumab for patients with advanced RCC following treatment with antiangiogenic therapy based on improved overall survival compared with the standard of care. Ongoing phase III trials are comparing checkpoint-inhibitor-based combination regimens with antiangiogenesis agents in the first-line setting. The field is evolving rapidly, with many clinical trials already testing several checkpoint inhibitors alone, in combination, or with other targeted therapies. In addition, different novel immune therapies are being investigated including vaccines, T-cell agonists, and chimeric antigen receptor T cells. Determining which patients will benefit from these therapies and which combination approaches will result in better response will be important as this field evolves.
Collapse
Affiliation(s)
- Maria I Carlo
- Memorial Sloan Kettering Cancer Center 1275 York Avenue New York, New York 10065, USA
| | - Martin H Voss
- Memorial Sloan Kettering Cancer Center 1275 York Avenue New York, New York 10065, USA
| | - Robert J Motzer
- Memorial Sloan Kettering Cancer Center 1275 York Avenue New York, New York 10065, USA
| |
Collapse
|
46
|
Bennati C, Paglialunga L, Ricciuti B, Metro G, Marcomigni L, Gili A, Crinò L. Targeting EGFR and ALK in NSCLC: current evidence and future perspective. Lung Cancer Manag 2016; 5:79-90. [PMID: 30643552 PMCID: PMC6310340 DOI: 10.2217/lmt-2016-0005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/12/2016] [Indexed: 12/26/2022] Open
Abstract
The advent of molecular therapy targeting specific driver oncogenes has dramatically changed the prognosis of a subset of NSCLC, dilating survival and improving the quality of life of patients with advanced disease. Two of the major targets for treatment with receptor TKIs are the activated mutated forms of the EGFR and the ALK gene fusions. In advanced NSCLC patients harboring EGFR mutations or ALK rearrangements, the use of TKIs in the first-line setting, have provided unexpected large progression-free survival and overall survival benefits, compared with cytotoxic chemotherapy. However, despite initial responses and durable remissions, the development of resistance inevitably leads to treatment failure. The aim of this review is to discuss the treatment strategy currently used for tumors harboring these two genetic targets and to focus on what will be available in clinical practice in the near future.
Collapse
Affiliation(s)
- Chiara Bennati
- Department of Medical Oncology, S Maria della Misericordia Hospital, Perugia/Italy
| | - Luca Paglialunga
- Department of Medical Oncology, S Maria della Misericordia Hospital, Perugia/Italy
| | - Biagio Ricciuti
- Department of Medical Oncology, S Maria della Misericordia Hospital, Perugia/Italy
| | - Giulio Metro
- Department of Medical Oncology, S Maria della Misericordia Hospital, Perugia/Italy
| | - Luca Marcomigni
- Department of Medical Oncology, S Maria della Misericordia Hospital, Perugia/Italy
| | - Alessio Gili
- Department of Experimental Medicine, S Maria della Misericordia Hospital, Perugia/Italy
| | - Lucio Crinò
- Department of Medical Oncology, S Maria della Misericordia Hospital, Perugia/Italy
| |
Collapse
|
47
|
Lee SH, Danishmalik SN, Sin JI. DNA vaccines, electroporation and their applications in cancer treatment. Hum Vaccin Immunother 2016; 11:1889-900. [PMID: 25984993 DOI: 10.1080/21645515.2015.1035502] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Numerous animal studies and recent clinical studies have shown that electroporation-delivered DNA vaccines can elicit robust Ag-specific CTL responses and reduce disease severity. However, cancer antigens are generally poorly immunogenic, requiring special conditions for immune response induction. To date, many different approaches have been used to elicit Ag-specific CTL and anti-neoplastic responses to DNA vaccines against cancer. In vivo electroporation is one example, whereas others include DNA manipulation, xenogeneic antigen use, immune stimulatory molecule and immune response regulator application, DNA prime-boost immunization strategy use and different DNA delivery methods. These strategies likely increase the immunogenicity of cancer DNA vaccines, thereby contributing to cancer eradication. However, cancer cells are heterogeneous and might become CTL-resistant. Thus, understanding the CTL resistance mechanism(s) employed by cancer cells is critical to develop counter-measures for this immune escape. In this review, the use of electroporation as a DNA delivery method, the strategies used to enhance the immune responses, the cancer antigens that have been tested, and the escape mechanism(s) used by tumor cells are discussed, with a focus on the progress of clinical trials using cancer DNA vaccines.
Collapse
Key Words
- AFP, α-fetoprotein
- APCs, antigen presenting cells
- CEA, carcinoembryonic antigen
- CTLA-4, cytotoxic T lymphocyte-associated antigen-4
- DCs, dendritic cells
- DNA vaccine
- EP, electroporation
- GITR, glucocorticoid-induced tumor necrosis factor receptor family-related gene
- HPV, human papillomavirus
- HSP, heat shock protein
- HSV, herpes simplex virus
- ID, intradermal
- IM, intramuscular
- MAGE, melanoma-associated antigen
- MART, melanoma antigen recognized by T cells
- PAP, prostatic acid phosphatase
- PD, programmed death
- PRAME, preferentially expressed antigen in melanoma
- PSA, prostate-specific antigen
- PSMA, prostate-specific membrane antigen
- WT1, Wilm's tumor
- anti-tumor immunity
- cancer
- hTERT, human telomerase reverse transcriptase
- tumor immune evasion
Collapse
Affiliation(s)
- Si-Hyeong Lee
- a BK21 Plus Graduate Program; Department of Microbiology ; School of Medicine; Kangwon National University ; Chuncheon , Gangwon-do , Korea
| | | | | |
Collapse
|
48
|
Timilsina U, Gaur R. Modulation of apoptosis and viral latency - an axis to be well understood for successful cure of human immunodeficiency virus. J Gen Virol 2016; 97:813-824. [PMID: 26764023 DOI: 10.1099/jgv.0.000402] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human immunodeficiency virus (HIV) is the causative agent of the deadly disease AIDS, which is characterized by the progressive decline of CD4(+)T-cells. HIV-1-encoded proteins such as envelope gp120 (glycoprotein gp120), Tat (trans-activator of transcription), Nef (negative regulatory factor), Vpr (viral protein R), Vpu (viral protein unique) and protease are known to be effective in modulating host cell signalling pathways that lead to an alteration in apoptosis of both HIV-infected and uninfected bystander cells. Depending on the stage of the virus life cycle and host cell type, these viral proteins act as mediators of pro- or anti-apoptotic signals. HIV latency in viral reservoirs is a persistent phenomenon that has remained beyond the control of the human immune system. To cure HIV infections completely, it is crucial to reactivate latent HIV from cellular pools and to drive these apoptosis-resistant cells towards death. Several previous studies have reported the role of HIV-encoded proteins in apoptosis modulation, but the molecular basis for apoptosis evasion of some chronically HIV-infected cells and reactivated latently HIV-infected cells still needs to be elucidated. The current review summarizes our present understanding of apoptosis modulation in HIV-infected cells, uninfected bystander cells and latently infected cells, with a focus on highlighting strategies to activate the apoptotic pathway to kill latently infected cells.
Collapse
Affiliation(s)
- Uddhav Timilsina
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi- 110021, India
| | - Ritu Gaur
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi- 110021, India
| |
Collapse
|
49
|
Brunn ND, Mauze S, Gu D, Wiswell D, Ueda R, Hodges D, Beebe AM, Zhang S, Escandon E. The Role of Anti-Drug Antibodies in the Pharmacokinetics, Disposition, Target Engagement, and Efficacy of a GITR Agonist Monoclonal Antibody in Mice. ACTA ACUST UNITED AC 2015; 356:574-86. [DOI: 10.1124/jpet.115.229864] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/14/2015] [Indexed: 12/17/2022]
|
50
|
Targeting the Immune System for Cancer Therapy: Lessons for Perioperative Management? CURRENT ANESTHESIOLOGY REPORTS 2015. [DOI: 10.1007/s40140-015-0111-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|