1
|
Santollani L, Wittrup KD. Spatiotemporally programming cytokine immunotherapies through protein engineering. Immunol Rev 2023; 320:10-28. [PMID: 37409481 DOI: 10.1111/imr.13234] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Cytokines have long been considered promising cancer immunotherapy agents due to their endogenous role in activating and proliferating lymphocytes. However, since the initial FDA approvals of Interleukin-2 (IL-2) and Interferon-ɑ (IFNɑ) for oncology over 30 years ago, cytokines have achieved little success in the clinic due to narrow therapeutic windows and dose-limiting toxicities. This is attributable to the discrepancy between the localized, regulated manner in which cytokines are deployed endogenously versus the systemic, untargeted administration used to date in most exogenous cytokine therapies. Furthermore, cytokines' ability to stimulate multiple cell types, often with paradoxical effects, may present significant challenges for their translation into effective therapies. Recently, protein engineering has emerged as a tool to address the shortcomings of first-generation cytokine therapies. In this perspective, we contextualize cytokine engineering strategies such as partial agonism, conditional activation and intratumoral retention through the lens of spatiotemporal regulation. By controlling the time, place, specificity, and duration of cytokine signaling, protein engineering can allow exogenous cytokine therapies to more closely approach their endogenous exposure profile, ultimately moving us closer to unlocking their full therapeutic potential.
Collapse
Affiliation(s)
- Luciano Santollani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - K Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
2
|
Rolfo C, Giovannetti E, Martinez P, McCue S, Naing A. Applications and clinical trial landscape using Toll-like receptor agonists to reduce the toll of cancer. NPJ Precis Oncol 2023; 7:26. [PMID: 36890302 PMCID: PMC9995514 DOI: 10.1038/s41698-023-00364-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 02/17/2023] [Indexed: 03/10/2023] Open
Abstract
Toll-like receptors (TLRs), which serve as a bridge between innate and adaptive immunity, may be viable treatment targets. TLRs are the first line of defense against microbes and activate signaling cascades that induce immune and inflammatory responses. Patients with "hot" versus "cold" tumors may respond more favorably to immune checkpoint inhibition, and through their downstream effects, TLR agonists have the potential to convert "cold tumors" into "hot tumors" making TLRs in combination with immune checkpoint inhibitors, potential targets for cancer therapies. Imiquimod is a topical TLR7 agonist, approved by the FDA for antiviral and skin cancer treatments. Other TLR adjuvants are used in several vaccines including Nu Thrax, Heplisav, T-VEC, and Cervarix. Many TLR agonists are currently in development as both monotherapy and in combination with immune checkpoint inhibitors. In this review, we describe the TLR agonists that are being evaluated clinically as new therapies for solid tumors.
Collapse
Affiliation(s)
- Christian Rolfo
- Center for Thoracic Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, NY, USA.
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands.,Cancer Pharmacology Lab, AIRC Start-Up unit, Fondazione Pisana per la Scienza, Pisa, Italy
| | | | | | - Aung Naing
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
3
|
Knight A, Karapetyan L, Kirkwood JM. Immunotherapy in Melanoma: Recent Advances and Future Directions. Cancers (Basel) 2023; 15:1106. [PMID: 36831449 PMCID: PMC9954703 DOI: 10.3390/cancers15041106] [Citation(s) in RCA: 61] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
The use of immunotherapy in the treatment of advanced and high-risk melanoma has led to a striking improvement in outcomes. Although the incidence of melanoma has continued to rise, median survival has improved from approximately 6 months to nearly 6 years for patients with advanced inoperable stage IV disease. Recent understanding of the tumor microenvironment and its interplay with the immune system has led to the explosive development of novel immunotherapy treatments. Since the approval of the therapeutic cytokines interleukin-2 and interferon alfa-2 in the 1990s, the development of novel immune checkpoint inhibitors (ICIs), oncolytic virus therapy, and modulators of the tumor microenvironment have given way to a new era in melanoma treatment. Monoclonal antibodies directed at programmed cell death protein 1 receptor (PD-1) and its ligand (PDL-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and lymphocyte-activation gene 3 (LAG-3) have provided robust activation of the adaptive immune system, restoring immune surveillance leading to host tumor recognition and destruction. Multiple other immunomodulatory therapeutics are under investigation to overcome resistance to ICI therapy, including the toll-like receptor-9 (TLR-9) and 7/8 (TLR-7/8) agonists, stimulator of interferon genes (STING) agonists, and fecal microbiota transplantation. In this review, we focus on the recent advances in immunotherapy for the treatment of melanoma and provide an update on novel therapies currently under investigation.
Collapse
Affiliation(s)
- Andrew Knight
- Department of Medicine, Division of General Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Lilit Karapetyan
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - John M. Kirkwood
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
4
|
Carvajal RD, Sacco JJ, Jager MJ, Eschelman DJ, Olofsson Bagge R, Harbour JW, Chieng ND, Patel SP, Joshua AM, Piperno-Neumann S. Advances in the clinical management of uveal melanoma. Nat Rev Clin Oncol 2023; 20:99-115. [PMID: 36600005 DOI: 10.1038/s41571-022-00714-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 01/05/2023]
Abstract
Melanomas arising in the uveal tract of the eye are a rare form of the disease with a biology and clinical phenotype distinct from their more common cutaneous counterparts. Treatment of primary uveal melanoma with radiotherapy, enucleation or other modalities achieves local control in more than 90% of patients, although 40% or more ultimately develop distant metastases, most commonly in the liver. Until January 2022, no systemic therapy had received regulatory approval for patients with metastatic uveal melanoma, and these patients have historically had a dismal prognosis owing to the limited efficacy of the available treatments. A series of seminal studies over the past two decades have identified highly prevalent early, tumour-initiating oncogenic genomic aberrations, later recurring prognostic alterations and immunological features that characterize uveal melanoma. These advances have driven the development of a number of novel emerging treatments, including tebentafusp, the first systemic therapy to achieve regulatory approval for this disease. In this Review, our multidisciplinary and international group of authors summarize the biology of uveal melanoma, management of primary disease and surveillance strategies to detect recurrent disease, and then focus on the current standard and emerging regional and systemic treatment approaches for metastatic uveal melanoma.
Collapse
Affiliation(s)
- Richard D Carvajal
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| | - Joseph J Sacco
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - David J Eschelman
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - J William Harbour
- Department of Ophthalmology and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Nicholas D Chieng
- Medical Imaging Services, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Sapna P Patel
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Anthony M Joshua
- Department of Medical Oncology, Kinghorn Cancer Centre, St Vincent's Hospital Sydney and Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,School of Clinical Medicine, UNSW Medicine & Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW, Sydney, New South Wales, Australia
| | | |
Collapse
|
5
|
Punekar SR, Weber JS. Intratumoral Therapy to Make a "Cold" Tumor "Hot": The Jury Is Still Out. Clin Cancer Res 2022; 28:5007-5009. [PMID: 36161479 DOI: 10.1158/1078-0432.ccr-22-2427] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 01/24/2023]
Abstract
Tilsotolimod, an oligodeoxynucleotide TLR9 agonist, administered intratumorally, has been clinically evaluated. This compound has demonstrated the ability to induce changes within the tumor microenvironment, to convert noninflamed cold tumors into inflamed hot tumors, with the hope that these tumors will be more responsive to immune checkpoint blockade. See related article by Babiker et al., p. 5079.
Collapse
Affiliation(s)
- Salman R Punekar
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York
| | - Jeffrey S Weber
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York
| |
Collapse
|
6
|
Zúñiga LA, Leßmann T, Uppal K, Bisek N, Hong E, Rasmussen CE, Karlsson JJ, Zettler J, Holten-Andersen L, Bang K, Thakar D, Lee YC, Martinez S, Sabharwal SS, Stark S, Faltinger F, Kracker O, Weisbrod S, Müller R, Voigt T, Bigott K, Tabrizifard M, Breinholt VM, Mirza AM, Rosen DB, Sprogøe K, Punnonen J. Intratumoral delivery of TransCon ™ TLR7/8 Agonist promotes sustained anti-tumor activity and local immune cell activation while minimizing systemic cytokine induction. Cancer Cell Int 2022; 22:286. [PMID: 36123697 PMCID: PMC9484246 DOI: 10.1186/s12935-022-02708-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022] Open
Abstract
Background Intratumoral (IT) delivery of toll-like receptor (TLR) agonists has shown encouraging anti-tumor benefit in preclinical and early clinical studies. However, IT delivery of TLR agonists may lead to rapid effusion from the tumor microenvironment (TME), potentially limiting the duration of local inflammation and increasing the risk of systemic adverse events. Methods To address these limitations, TransCon™ TLR7/8 Agonist—an investigational sustained-release prodrug of resiquimod that uses a TransCon linker and hydrogel technology to achieve sustained and predictable IT release of resiquimod—was developed. TransCon TLR7/8 Agonist was characterized for resiquimod release in vitro and in vivo, in mice and rats, and was assessed for anti-tumor efficacy and pharmacodynamic activity in mice. Results Following a single IT dose, TransCon TLR7/8 Agonist mediated potent tumor growth inhibition which was associated with sustained resiquimod release over several weeks with minimal induction of systemic cytokines. TransCon TLR7/8 Agonist monotherapy promoted activation of antigen-presenting cells in the TME and tumor-draining lymph nodes, with evidence of activation and expansion of CD8+ T cells in the tumor-draining lymph node and TME. Combination of TransCon TLR7/8 Agonist with systemic immunotherapy further promoted anti-tumor activity in TransCon TLR7/8 Agonist-treated tumors. In a bilateral tumor setting, combination of TransCon TLR7/8 Agonist with systemic IL-2 potentiated tumor growth inhibition in both injected and non-injected tumors and conferred protection against tumor rechallenge following complete regressions. Conclusions Our findings show that a single dose of TransCon TLR7/8 Agonist can mediate sustained local release of resiquimod in the TME and promote potent anti-tumor effects as monotherapy and in combination with systemic immunotherapy, supporting TransCon TLR7/8 Agonist as a novel intratumoral TLR agonist for cancer therapy. A clinical trial to evaluate the safety and efficacy of TransCon TLR7/8 Agonist, as monotherapy and in combination with pembrolizumab, in cancer patients is currently ongoing (transcendIT-101; NCT04799054). Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02708-6.
Collapse
Affiliation(s)
| | | | - Karan Uppal
- Ascendis Pharma, Inc., Redwood City, CA, USA
| | | | - Enping Hong
- Ascendis Pharma, Inc., Redwood City, CA, USA
| | | | | | | | | | - Kathy Bang
- Ascendis Pharma, Inc., Redwood City, CA, USA
| | | | - Yu-Chi Lee
- Ascendis Pharma, Inc., Redwood City, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Marconcini R, Pezzicoli G, Stucci LS, Sergi MC, Lospalluti L, Porta C, Tucci M. Combination of immunotherapy and other targeted therapies in advanced cutaneous melanoma. Hum Vaccin Immunother 2022; 18:1980315. [PMID: 34613889 PMCID: PMC9302493 DOI: 10.1080/21645515.2021.1980315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 08/22/2021] [Accepted: 09/08/2021] [Indexed: 12/20/2022] Open
Abstract
Cutaneous Melanoma (CM) is an aggressive cancer whose incidence is increasing worldwide. However, the knowledge of its biology and genes driving cell growth and survival allowed to develop new drugs that have improved PFS and OS of advanced disease. Both BRAF targeting agents and immune checkpoint inhibitors (ICIs) have been adopted for the treatment of metastatic disease and the adjuvant setting. Several melanoma patients show innate or acquired drug-resistance and thus new strategies are required for overcoming this complication. New ICIs have been developed, and strategies of combination or sequencing are under investigation in ongoing clinical trials. In addition, pre-clinical data have demonstrated that many strategies induce the release of neoantigens within the tumor microenvironment, thus suggesting the combination of new agents with ICIs. Here, we review the ongoing strategies in advanced CM including a dedicated section on treatment of brain metastases.
Collapse
Affiliation(s)
- Riccardo Marconcini
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Gaetano Pezzicoli
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Policlinico di Bari, Bari, Italy
- Dermatology Unit, Azienda Ospedaliero-Universitaria Policlinico di Bari, Bari, Italy
| | - Luigia Stefania Stucci
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Policlinico di Bari, Bari, Italy
| | - Maria Chiara Sergi
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Policlinico di Bari, Bari, Italy
| | - Lucia Lospalluti
- Dermatology Unit, Azienda Ospedaliero-Universitaria Policlinico di Bari, Bari, Italy
| | - Camillo Porta
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Policlinico di Bari, Bari, Italy
- Department of Biomedical Sciences and Human Oncolog, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Marco Tucci
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Policlinico di Bari, Bari, Italy
- Department of Biomedical Sciences and Human Oncolog, University of Bari ‘Aldo Moro’, Bari, Italy
| |
Collapse
|
8
|
DePalo DK, Tarhini A, Zager JS. The treatment of advanced melanoma: a review of systemic and local therapies in combination with immune checkpoint inhibitors in phase 1 and 2 clinical trials. Expert Opin Investig Drugs 2022; 31:95-104. [PMID: 34996314 DOI: 10.1080/13543784.2022.2027366] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION While the incidence of melanoma continues to rise, the mortality of the disease appears to have stabilized. This may, in part, be due to the development and application of immune checkpoint inhibitors as standard of care in advanced melanoma. However, many patients do not respond to these therapies alone. Combining immune checkpoint inhibitors with other classes of therapeutics appears to be a promising direction to improve response and survival in advanced melanoma. AREAS COVERED This review article aims to discuss phase 1 and 2 clinical trials examining immune checkpoint inhibitors in combination therapy for the treatment of advanced, unresectable melanoma. In particular, these regimens include various kinase inhibitors, tumor-infiltrating lymphocytes, toll-like receptor agonists, cytokines, and oncolytic viral therapies. The combinations under discussion include both systemic and combination systemic/local therapies. EXPERT OPINION Drug combinations discussed here appear to be promising therapeutic regimens for advanced melanoma. Improved understanding of the mechanisms of primary, adaptive, and acquired resistance to immune checkpoint inhibitors may guide the development of future combination regimens.
Collapse
Affiliation(s)
- Danielle K DePalo
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Ahmad Tarhini
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
9
|
Hartimath SV, Ramasamy B, Xuan TY, Rong TJ, Khanapur S, Cheng P, Hwang YY, Robins EG, Goggi JL. Granzyme B PET Imaging in Response to In Situ Vaccine Therapy Combined with αPD1 in a Murine Colon Cancer Model. Pharmaceutics 2022; 14:pharmaceutics14010150. [PMID: 35057046 PMCID: PMC8779135 DOI: 10.3390/pharmaceutics14010150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/16/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) block checkpoint receptors that tumours use for immune evasion, allowing immune cells to target and destroy cancer cells. Despite rapid advancements in immunotherapy, durable response rates to ICIs remains low. To address this, combination clinical trials are underway assessing whether adjuvants can enhance responsiveness by increasing tumour immunogenicity. CpG-oligodeoxynucleotides (CpG-ODN) are synthetic DNA fragments containing an unmethylated cysteine-guanosine motif that stimulate the innate and adaptive immune systems by engaging Toll-like receptor 9 (TLR9) present on the plasmacytoid dendritic cells (pDCs) and B cells. Here, we have assessed the ability of AlF-mNOTA-GZP, a peptide tracer targeting granzyme B, to serve as a PET imaging biomarker in response to CpG-ODN 1585 in situ vaccine therapy delivered intratumourally (IT) or intraperitoneally (IP) either as monotherapy or in combination with αPD1. [18F]AlF-mNOTA-GZP was able to differentiate treatment responders from non-responders based on tumour uptake. Furthermore, [18F]AlF-mNOTA-GZP showed positive associations with changes in tumour-associated lymphocytes expressing GZB, namely GZB+ CD8+ T cells, and decreases in suppressive F4/80+ cells. [18F]AlF-mNOTA-GZP tumour uptake was mediated by GZB expressing CD8+ cells and successfully stratifies therapy responders from non-responders, potentially acting as a non-invasive biomarker for ICIs and combination therapy evaluation in a clinical setting.
Collapse
Affiliation(s)
- Siddesh V. Hartimath
- Laboratory of Radiochemistry & Molecular Imaging (LRMI), Institute of Bioengineering and Bioimaging (IBB), A*STAR Research Entities, Helios, Singapore 138667, Singapore; (B.R.); (T.Y.X.); (T.J.R.); (S.K.); (P.C.); (E.G.R.)
- Correspondence: (S.V.H.); (J.L.G.)
| | - Boominathan Ramasamy
- Laboratory of Radiochemistry & Molecular Imaging (LRMI), Institute of Bioengineering and Bioimaging (IBB), A*STAR Research Entities, Helios, Singapore 138667, Singapore; (B.R.); (T.Y.X.); (T.J.R.); (S.K.); (P.C.); (E.G.R.)
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Tan Yun Xuan
- Laboratory of Radiochemistry & Molecular Imaging (LRMI), Institute of Bioengineering and Bioimaging (IBB), A*STAR Research Entities, Helios, Singapore 138667, Singapore; (B.R.); (T.Y.X.); (T.J.R.); (S.K.); (P.C.); (E.G.R.)
| | - Tang Jun Rong
- Laboratory of Radiochemistry & Molecular Imaging (LRMI), Institute of Bioengineering and Bioimaging (IBB), A*STAR Research Entities, Helios, Singapore 138667, Singapore; (B.R.); (T.Y.X.); (T.J.R.); (S.K.); (P.C.); (E.G.R.)
| | - Shivashankar Khanapur
- Laboratory of Radiochemistry & Molecular Imaging (LRMI), Institute of Bioengineering and Bioimaging (IBB), A*STAR Research Entities, Helios, Singapore 138667, Singapore; (B.R.); (T.Y.X.); (T.J.R.); (S.K.); (P.C.); (E.G.R.)
| | - Peter Cheng
- Laboratory of Radiochemistry & Molecular Imaging (LRMI), Institute of Bioengineering and Bioimaging (IBB), A*STAR Research Entities, Helios, Singapore 138667, Singapore; (B.R.); (T.Y.X.); (T.J.R.); (S.K.); (P.C.); (E.G.R.)
| | - You Yi Hwang
- FACS facility, Singapore Immunology Network (SIgN), A*STAR Research Entities, Immunos, Singapore 138665, Singapore;
| | - Edward G. Robins
- Laboratory of Radiochemistry & Molecular Imaging (LRMI), Institute of Bioengineering and Bioimaging (IBB), A*STAR Research Entities, Helios, Singapore 138667, Singapore; (B.R.); (T.Y.X.); (T.J.R.); (S.K.); (P.C.); (E.G.R.)
- Clinical Imaging Research Centre (CIRC), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Julian L. Goggi
- Laboratory of Radiochemistry & Molecular Imaging (LRMI), Institute of Bioengineering and Bioimaging (IBB), A*STAR Research Entities, Helios, Singapore 138667, Singapore; (B.R.); (T.Y.X.); (T.J.R.); (S.K.); (P.C.); (E.G.R.)
- Correspondence: (S.V.H.); (J.L.G.)
| |
Collapse
|
10
|
Comito F, Pagani R, Grilli G, Sperandi F, Ardizzoni A, Melotti B. Emerging Novel Therapeutic Approaches for Treatment of Advanced Cutaneous Melanoma. Cancers (Basel) 2022; 14:271. [PMID: 35053435 PMCID: PMC8773625 DOI: 10.3390/cancers14020271] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/29/2021] [Accepted: 01/04/2022] [Indexed: 02/01/2023] Open
Abstract
The prognosis of patients with advanced cutaneous melanoma has radically changed in the past decade. Nevertheless, primary or acquired resistance to systemic treatment occurs in many cases, highlighting the need for novel treatment strategies. This review has the purpose of summarizing the current area of interest for the treatment of metastatic or unresectable advanced cutaneous melanoma, including data from recently completed or ongoing clinical trials. The main fields of investigation include the identification of new immune checkpoint inhibitors (anti-LAG3, GITR agonist and anti-TIGIT), adoptive cell therapy, vaccines, engineered TCR therapy, IL-2 agonists, novel targets for targeted therapy (new MEK or RAF inhibitors, HDAC, IDO, ERK, Axl, ATR and PARP inhibitors), or combination strategies (antiangiogenetic agents plus immune checkpoint inhibitors, intra-tumoral immunotherapy in combination with systemic therapy). In many cases, only preliminary efficacy data from early phase trials are available, which require confirmation in larger patient cohorts. A more in-depth knowledge of the biological effects of the molecules and identifying predictive biomarkers remain crucial for selecting patient populations most likely to benefit from novel emerging treatment strategies.
Collapse
Affiliation(s)
- Francesca Comito
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni, 15-40138 Bologna, Italy; (G.G.); (F.S.); (A.A.); (B.M.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti, 9-40138 Bologna, Italy
| | - Rachele Pagani
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni, 15-40138 Bologna, Italy; (G.G.); (F.S.); (A.A.); (B.M.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti, 9-40138 Bologna, Italy
| | - Giada Grilli
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni, 15-40138 Bologna, Italy; (G.G.); (F.S.); (A.A.); (B.M.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti, 9-40138 Bologna, Italy
| | - Francesca Sperandi
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni, 15-40138 Bologna, Italy; (G.G.); (F.S.); (A.A.); (B.M.)
| | - Andrea Ardizzoni
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni, 15-40138 Bologna, Italy; (G.G.); (F.S.); (A.A.); (B.M.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti, 9-40138 Bologna, Italy
| | - Barbara Melotti
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni, 15-40138 Bologna, Italy; (G.G.); (F.S.); (A.A.); (B.M.)
| |
Collapse
|
11
|
Mehmi I, Hamid O. Immunotherapy of cancer in the era of checkpoint inhibitor. Clin Exp Metastasis 2021; 39:231-237. [PMID: 34878618 DOI: 10.1007/s10585-021-10132-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 11/02/2021] [Indexed: 12/15/2022]
Abstract
Application of immunotherapy has revolutionized treatment of number of malignancies. We present a review of immunotherapy approaches, early-phase data of number of new immunotherapeutic targets in melanoma, cutaneous squamous cell carcinoma, Merkel cell cancer, and non-small cell lung cancer.
Collapse
Affiliation(s)
- Inderjit Mehmi
- The Angeles Clinic and Research Institute, Cedar Sinai Affiliate, Los Angeles, CA, USA.
| | - Omid Hamid
- The Angeles Clinic and Research Institute, Cedar Sinai Affiliate, Los Angeles, CA, USA
| |
Collapse
|
12
|
A Squalene-Based Nanoemulsion for Therapeutic Delivery of Resiquimod. Pharmaceutics 2021; 13:pharmaceutics13122060. [PMID: 34959344 PMCID: PMC8706843 DOI: 10.3390/pharmaceutics13122060] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 12/03/2022] Open
Abstract
Agonists for toll-like receptors (TLRs) have shown promising activities against cancer. In the present study, a squalene-based nanoemulsion (NE) was loaded with resiquimod, a TLR7/8 agonist for therapeutic delivery. R848 NE was developed and characterized for long-term stability. In vitro and in vivo antitumor immunity of R848 NE were also evaluated in combination with SD-101, a CpG-containing TLR9 agonist. In vitro studies demonstrated strong long-term stability and immune responses to R848 NE. When combined with SD-101, strong antitumor activity was observed in MC38 murine colon carcinoma model with over 80% tumor growth inhibition. The combination treatment showed a 4-fold increase in systemic TNFa production and a 2.6-fold increase in Cd8a expression in tumor tissues, suggesting strong cell-mediated immune responses against the tumor. The treatment not only demonstrated a strong antitumor immunity by TLR7/8 and TLR9 activations but also induced PD-L1 upregulation in tumors, suggesting a potential therapeutic synergy with immune checkpoint inhibitors.
Collapse
|
13
|
Huppert LA, Mariotti V, Chien AJ, Soliman HH. Emerging immunotherapeutic strategies for the treatment of breast cancer. Breast Cancer Res Treat 2021; 191:243-255. [PMID: 34716870 DOI: 10.1007/s10549-021-06406-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022]
Abstract
Immunotherapy has resulted in unprecedented gains in long-term outcomes for many cancer types and has revolutionized the treatment landscape of solid tumor oncology. Checkpoint inhibition in combination with chemotherapy has proven to be effective for the treatment of a subset of advanced triple-negative breast cancer in the first-line setting. This initial success is likely just the tip of the iceberg as there is much that remains unknown about how to best harness the immune system as a therapeutic strategy in all breast cancer subtypes. Therefore, numerous ongoing studies are currently underway to evaluate the safety and efficacy of immunotherapy in breast cancer. In this review, we will discuss emerging immunotherapeutic strategies for breast cancer treatment including the following: (1) Intratumoral therapies, (2) Anti-tumor vaccines, (3) B-specific T-cell engagers, and (4) Chimeric antigen receptor T-cell therapy, and (5) Emerging systemic immunotherapy strategies. For each topic, we will review the existing preclinical and clinical literature, discuss ongoing clinical trials, and highlight future directions in the field.
Collapse
Affiliation(s)
- Laura A Huppert
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | | | - A Jo Chien
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Hatem H Soliman
- Department of Breast Oncology, H Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| |
Collapse
|
14
|
Gerard CL, Delyon J, Wicky A, Homicsko K, Cuendet MA, Michielin O. Turning tumors from cold to inflamed to improve immunotherapy response. Cancer Treat Rev 2021; 101:102227. [PMID: 34656019 DOI: 10.1016/j.ctrv.2021.102227] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 05/13/2021] [Accepted: 05/15/2021] [Indexed: 12/30/2022]
Abstract
Immune checkpoint inhibitors have revolutionized the treatment landscape for a number of cancers over the last few decades. Nevertheless, a majority of patients still do not benefit from these treatments. Such patient-specific lack of response can be predicted, in part, from the immune phenotypes present in the tumor microenvironment. We provide a perspective on options to reprogram the tumors and their microenvironment to increase the therapeutic efficacy of immunotherapies and expand their efficacy against cold tumors. Additionally, we review data from current preclinical and clinical trials aimed at testing the different therapeutic options in monotherapy or preferably in combination with checkpoint inhibitors.
Collapse
Affiliation(s)
- C L Gerard
- Precision Oncology Center, Lausanne University Hospital (CHUV), Switzerland
| | - J Delyon
- Precision Oncology Center, Lausanne University Hospital (CHUV), Switzerland
| | - A Wicky
- Precision Oncology Center, Lausanne University Hospital (CHUV), Switzerland
| | - K Homicsko
- Precision Oncology Center, Lausanne University Hospital (CHUV), Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Michel A Cuendet
- Precision Oncology Center, Lausanne University Hospital (CHUV), Switzerland; Molecular Modelling Group, Swiss Institute of Bioinformatics, Lausanne, Switzerland; Department of Physiology and Biophysics, Weill Cornell Medicine, NY, USA.
| | - O Michielin
- Precision Oncology Center, Lausanne University Hospital (CHUV), Switzerland; Molecular Modelling Group, Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| |
Collapse
|
15
|
Yuan J, Khilnani A, Brody J, Andtbacka RHI, Hu-Lieskovan S, Luke JJ, Diab A, Marabelle A, Snyder A, Cao ZA, Hodi FS. Current strategies for intratumoural immunotherapy - Beyond immune checkpoint inhibition. Eur J Cancer 2021; 157:493-510. [PMID: 34561127 DOI: 10.1016/j.ejca.2021.08.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/26/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022]
Abstract
Immunotherapy has revolutionised cancer treatment through restoration of host antitumour immune response. Immune checkpoint inhibitors (ICIs) confer durable responses in only a subset of patients. Mechanisms of ICI resistance to improve durable response rates and overall survival are an area of intense clinical research. Robust clinical development is ongoing to evaluate novel combination therapies to overcome ICI resistance, including targeting immunoregulatory pathways in the tumour microenvironment. Intratumoural (IT) immunotherapies such as toll-like receptor agonists, stimulator of interferon-induced gene agonists, retinoic-inducible gene I-like receptor agonists and oncolytic viruses may represent potential combination treatment options to overcome ICI resistance. Use of IT immunotherapies in combination with ICIs may alter the tumour microenvironment to address resistance mechanisms and improve antitumour response. Optimisation of IT immunotherapy clinical trials will elucidate resistance mechanisms, facilitate clinical trial design, define pharmacodynamic predictors that identify patients who may most benefit and inform clinical development of combination immunotherapy regimens. Here we provide an overview of IT immunotherapy principles, mechanisms of action, categories of IT immunotherapeutics, emerging data, clinical development strategies, response assessment, dose and schedule determination, clinical trial design and translational study design.
Collapse
Affiliation(s)
- Jianda Yuan
- Department of Translational Oncology and Early Oncology Development, Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA.
| | - Anuradha Khilnani
- Department of Translational Oncology and Early Oncology Development, Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA.
| | - Joshua Brody
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.
| | - Robert H I Andtbacka
- Seven and Eight Biopharmaceuticals Inc., 343 Thornall Street, Suite 520, Edison, NJ, 08837, USA.
| | - Siwen Hu-Lieskovan
- Department of Internal Medicine-Oncology, Huntsman Cancer Institute, University of Utah, 2000 Cir of Hope Dr #1950, Salt Lake City, UT, 84112, USA.
| | - Jason J Luke
- Department of Hematology/Oncology, University of Pittsburgh Hillman Cancer Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA.
| | - Adi Diab
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Aurelien Marabelle
- Department of Therapeutic Innovation and Early Trials, Gustave Roussy, University of Paris-Saclay, 114 Rue Edouard Vaillant, 94805 Villejuif, France.
| | - Alexandra Snyder
- Department of Translational Oncology and Early Oncology Development, Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA.
| | - Z Alexander Cao
- Department of Translational Oncology and Early Oncology Development, Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA.
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA.
| |
Collapse
|
16
|
Saeed M, Chen F, Ye J, Shi Y, Lammers T, De Geest BG, Xu ZP, Yu H. From Design to Clinic: Engineered Nanobiomaterials for Immune Normalization Therapy of Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2008094. [PMID: 34048101 DOI: 10.1002/adma.202008094] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/17/2021] [Indexed: 05/21/2023]
Abstract
The tumor immune microenvironment (TIME) is comprised of a complex milieu that contributes to stunting antitumor immune responses by restricting T cells to accumulate in the vicinity of the tumor. Nanomedicine-based strategies are being proposed as a salvage effort to reinvigorate antitumor immunity. Various strategies, however, often fail to unleash the antitumor immune response because of the paucity of appropriate therapeutic targets in the complex TIME, invigorating a fervor of investigation into mechanisms underlying the TIME to resist nanomedicines. In this review article, effective nano/biomaterial-based delivery and TIME normalization approaches that promote T cell-mediated antitumor immune response will be discussed, with a focus on emerging preclinical and clinical strategies for immune normalization. Based on currently available evidence, it seems as if the ultimate success of cancer immunotherapy and nanomedicine hinges on the capacity to normalize the TIME. Here, how nanomedicines target immunosuppressive cells and signaling pathways to broaden the impact of cancer immunotherapy are explored. Acquisition of the urgently needed knowledge of nanomedicine-mediated immune normalization will guide researchers and scientists towards clinical applications of cancer immunotherapy.
Collapse
Affiliation(s)
- Madiha Saeed
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Fangming Chen
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Jiayi Ye
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Yang Shi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany
| | - Bruno G De Geest
- Department of Pharmaceutics and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, 9000, Belgium
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| |
Collapse
|
17
|
Trojaniello C, Luke JJ, Ascierto PA. Therapeutic Advancements Across Clinical Stages in Melanoma, With a Focus on Targeted Immunotherapy. Front Oncol 2021; 11:670726. [PMID: 34178657 PMCID: PMC8222774 DOI: 10.3389/fonc.2021.670726] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/10/2021] [Indexed: 12/31/2022] Open
Abstract
Melanoma is the most fatal skin cancer. In the early stages, it can be safely treated with surgery alone. However, since 2011, there has been an important revolution in the treatment of melanoma with new effective treatments. Targeted therapy and immunotherapy with checkpoint inhibitors have changed the history of this disease. To date, more than half of advanced melanoma patients are alive at 5 years; despite this breakthrough, approximately half of the patients still do not respond to treatment. For these reasons, new therapeutic strategies are required to expand the number of patients who can benefit from immunotherapy or combination with targeted therapy. Current research aims at preventing primary and acquired resistance, which are both responsible for treatment failure in about 50% of patients. This could increase the effectiveness of available drugs and allow for the evaluation of new combinations and new targets. The main pathways and molecules under study are the IDO inhibitor, TLR9 agonist, STING, LAG-3, TIM-3, HDAC inhibitors, pegylated IL-2 (NKTR-214), GITR, and adenosine pathway inhibitors, among others (there are currently about 3000 trials that are evaluating immunotherapeutic combinations in different tumors). Other promising strategies are cancer vaccines and oncolytic viruses. Another approach is to isolate and remove immune cells (DCs, T cells, and NK cells) from the patient's blood or tumor infiltrates, add specific gene fragments, expand them in culture with growth factors, and re-inoculate into the same patient. TILs, TCR gene transfer, and CAR-T therapy follow this approach. In this article, we give an overview over the current status of melanoma therapies, the clinical rationale for choosing treatments, and the new immunotherapy approaches.
Collapse
Affiliation(s)
- Claudia Trojaniello
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Jason J. Luke
- Cancer Immunotherapeutics Center, University of Pittsburgh Medical Center and Hillman Cancer Center, Pittsburgh, PA, United States
| | - Paolo A. Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| |
Collapse
|
18
|
Dang N, Waer M, Sprangers B, Lin Y. Intratumoral immunotherapy with anti-PD-1 and TLR9 agonist induces systemic antitumor immunity without accelerating rejection of cardiac allografts. Am J Transplant 2021; 21:60-72. [PMID: 32506732 DOI: 10.1111/ajt.16105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/21/2020] [Accepted: 05/23/2020] [Indexed: 01/25/2023]
Abstract
Immune checkpoint inhibitors, such as programmed cell death 1 (PD-1) blockades, have revolutionized the field of cancer immunotherapy. However, there is a growing concern whether PD-1 inhibitors can be administered safely to transplant recipients with advanced cancer, as the T cells activated by checkpoint inhibitors may become reactive not only toward tumor antigens but also toward donor alloantigen, thereby resulting in allograft rejection. Here, immunotherapy with anti-PD-1/toll like receptor 9 agonist was administered to C57BL/6 mice bearing a cardiac allograft that were receiving maintenance immunosuppression or a PI4KIIIβ inhibitor-based tolerogenic regimen. Intratumoral (i.t.), but not systemic, immunotherapy promoted potent anti-tumor responses, but did not accelerate allograft rejection. This effect was associated with a pro-immunogenic effect induced by i.t. immunotherapy resulting in systemic cellular and humoral immune anti-tumor responses. Furthermore, when the tumor and cardiac allograft shared major histocompatibility complex (MHC) antigens, i.t. immunotherapy promoted immune responses directed against tumor and the cardiac allograft resulting in allograft rejection. The anti-tumor effect was compromised by maintenance immunosuppression with cyclosporin A, indicating that an optimal balance between enhanced anti-tumor immunity and decreased transplant immunoreactivity is critical. A clinically relevant approach could be to temporarily withdraw maintenance immunosuppression and/or replace it with a PI4KIIIβ inhibitor-based tolerance-inducing regimen to allow for effective immunotherapy to take place.
Collapse
Affiliation(s)
- Nana Dang
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mark Waer
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Ben Sprangers
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.,Department of Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Yuan Lin
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Martin JD, Cabral H, Stylianopoulos T, Jain RK. Improving cancer immunotherapy using nanomedicines: progress, opportunities and challenges. Nat Rev Clin Oncol 2020; 17:251-266. [PMID: 32034288 DOI: 10.1038/s41571-019-0308-z] [Citation(s) in RCA: 378] [Impact Index Per Article: 94.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2019] [Indexed: 02/08/2023]
Abstract
Multiple nanotherapeutics have been approved for patients with cancer, but their effects on survival have been modest and, in some examples, less than those of other approved therapies. At the same time, the clinical successes achieved with immunotherapy have revolutionized the treatment of multiple advanced-stage malignancies. However, the majority of patients do not benefit from the currently available immunotherapies and many develop immune-related adverse events. By contrast, nanomedicines can reduce - but do not eliminate - the risk of certain life-threatening toxicities. Thus, the combination of these therapeutic classes is of intense research interest. The tumour microenvironment (TME) is a major cause of the failure of both nanomedicines and immunotherapies that not only limits delivery, but also can compromise efficacy, even when agents accumulate in the TME. Coincidentally, the same TME features that impair nanomedicine delivery can also cause immunosuppression. In this Perspective, we describe TME normalization strategies that have the potential to simultaneously promote the delivery of nanomedicines and reduce immunosuppression in the TME. Then, we discuss the potential of a combined nanomedicine-based TME normalization and immunotherapeutic strategy designed to overcome each step of the cancer-immunity cycle and propose a broadly applicable 'minimal combination' of therapies designed to increase the number of patients with cancer who are able to benefit from immunotherapy.
Collapse
Affiliation(s)
- John D Martin
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Human Anti-tumor Immunity: Insights from Immunotherapy Clinical Trials. Immunity 2020; 52:36-54. [DOI: 10.1016/j.immuni.2019.12.010] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/08/2019] [Accepted: 12/14/2019] [Indexed: 12/15/2022]
|
21
|
Leong WI, Ames RY, Haverkamp JM, Torres L, Kline J, Bans A, Rocha L, Gallotta M, Guiducci C, Coffman RL, Janatpour MJ. Low-dose metronomic cyclophosphamide complements the actions of an intratumoral C-class CpG TLR9 agonist to potentiate innate immunity and drive potent T cell-mediated anti-tumor responses. Oncotarget 2019; 10:7220-7237. [PMID: 31921384 PMCID: PMC6944447 DOI: 10.18632/oncotarget.27322] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/19/2019] [Indexed: 12/21/2022] Open
Abstract
The synthetic oligonucleotide SD-101 is a potent and specific agonist for toll-like receptor 9. Intratumoral injection of SD-101 induces significant anti-tumor immunity in preclinical and clinical studies, especially when combined with PD-1 blockade. To build upon this strategy, we studied the enhancement of SD-101 activities by combination with low-dose cyclophosphamide, a well-characterized agent with potentially complementary activities. In multiple mouse tumor models, we demonstrate substantial anti-tumor activity of the combination, compared to each single agent. Combination therapy generated CD8+ T cell dependent immunity leading to rejection of both non-injected and injected tumors and long-term survival, even in very large tumors. Mechanistic studies encompassing global gene expression changes and characterization of immune cell infiltrates show the rapid, sequential induction of innate and adaptive responses and identify discrete contributions of SD-101 and cyclophosphamide. Importantly, these changes were prominent in tumors not injected directly with SD-101. Combination treatment resulted in creation of a permissive environment for a systemic anti-tumor immune response, including a reduction of intratumoral regulatory T cells (Tregs) and an increase in "M1" versus "M2" tumor-associated macrophage (TAM) phenotypes. Additionally, we observed increased immunogenic cell death as well as antigen processing in response to combination treatment.
Collapse
Affiliation(s)
- Weng In Leong
- Dynavax Technologies, Inc., Emeryville, CA 94608, USA
| | | | | | - Laura Torres
- Dynavax Technologies, Inc., Emeryville, CA 94608, USA
| | - Janine Kline
- Dynavax Technologies, Inc., Emeryville, CA 94608, USA
| | - Ashil Bans
- Dynavax Technologies, Inc., Emeryville, CA 94608, USA
| | - Lauren Rocha
- Dynavax Technologies, Inc., Emeryville, CA 94608, USA
| | | | | | | | | |
Collapse
|
22
|
Jeong Y, Kim GB, Ji Y, Kwak GJ, Nam GH, Hong Y, Kim S, An J, Kim SH, Yang Y, Chung HS, Kim IS. Dendritic cell activation by an E. coli-derived monophosphoryl lipid A enhances the efficacy of PD-1 blockade. Cancer Lett 2019; 472:19-28. [PMID: 31857157 DOI: 10.1016/j.canlet.2019.12.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/03/2019] [Accepted: 12/06/2019] [Indexed: 12/30/2022]
Abstract
Cancer immunotherapy is a powerful approach for cancer treatment, but its clinical effects rely on the tumor's immune conditions. In particular, low response rates to PD-1 blockades are highly correlated with impaired T cell priming. Here, we demonstrate that E. coli-derived monophosphoryl lipid A (EcML) activates dendritic cells in a toll-like receptor-4 (TLR-4)-dependent manner and increases the sensitivity of cancer cells to anti-PD-1 immunotherapy. EcML is a mixture of 4'-monophosphoryl lipids A (MPLAs) produced directly by an engineered Escherichia coli strain; it has a unique congener composition that differentiates it from the well-established MPLA adjuvants, 3-O-desacyl-4'-monophosphoryl lipid A and glucopyranosyl lipid A. Given that active dendritic cells initiate adaptive immune responses, we investigated the anti-tumor activity of an aqueous formulation of EcML. Upon sensing EcML via TLR-4, dendritic cells matured into powerful antigen-presenting cells that could stimulate naïve T cells. EcML reduced tumor growth in the B16F10 mouse model via dendritic cell activation and potentiated PD-1 blockade therapy in the B16F10-OVA melanoma model. These data identify EcML as a promising TLR-4 agonist that can induce anti-tumor immune responses and potentiate PD-1 blockade therapy against tumors.
Collapse
Affiliation(s)
- Youngmin Jeong
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Gi Beom Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Yuhyun Ji
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Gi-Jung Kwak
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Gi-Hoon Nam
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Yeonsun Hong
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seohyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jinsu An
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Sun Hwa Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Yoosoo Yang
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Hak Suk Chung
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
23
|
Abstract
New therapies that promote antitumour immunity have been recently developed. Most of these immunomodulatory approaches have focused on enhancing T-cell responses, either by targeting inhibitory pathways with immune checkpoint inhibitors, or by targeting activating pathways, as with chimeric antigen receptor T cells or bispecific antibodies. Although these therapies have led to unprecedented successes, only a minority of patients with cancer benefit from these treatments, highlighting the need to identify new cells and molecules that could be exploited in the next generation of immunotherapy. Given the crucial role of innate immune responses in immunity, harnessing these responses opens up new possibilities for long-lasting, multilayered tumour control.
Collapse
|
24
|
Agrawal S, Kandimalla ER. Intratumoural immunotherapy: activation of nucleic acid sensing pattern recognition receptors. ACTA ACUST UNITED AC 2019; 3:15-23. [PMID: 35757301 PMCID: PMC9216656 DOI: 10.1016/j.iotech.2019.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Recently, it has become clear that the tumour microenvironment (TME) is important in cancer immunotherapy. While immune checkpoint inhibitors are effective for some patients, the heterogeneous nature and status of the TME (‘cold’ tumours) play a critical role in suppressing antitumour immunity in non-responding patients. Converting ‘cold’ to ‘hot’ tumours through modulation of the TME may enable expansion of the therapeutic efficacy of immunotherapy to a broader patient population. This paper describes advances in intratumoural immunotherapy, specifically activation of nucleic acid sensing pattern recognition receptors to modulate the TME. Intratumoural immunotherapy to modulate the tumour microenvironment. Use of novel immunostimulatory agents which activate nucleic acid sensing pattern recognition receptors. Harnessing innate and adaptive immunity induced by receptor-mediated immune cascade. Intratumoural therapy leads to local and anenestic tumour responses.
Collapse
Affiliation(s)
- Sudhir Agrawal
- University of Massachusetts Medical School, Department of Medicine, Worcester, USA
- ARNAY Sciences LLC, Shrewsbury, USA
- Corresponding author. Sudhir Agrawal, University of Massachusetts Medical School, Department of Medicine, 55 N Lake Ave, Worcester, MA 01655, USA.
| | | |
Collapse
|