1
|
Sergent P, Pinto-Cárdenas JC, Carrillo AJA, Dávalos DL, Pérez MDG, Lechuga DAM, Alonso-Miguel D, Schaafsma E, Cuarenta AJ, Muñoz DC, Zarabanda Y, Palisoul SM, Lewis PJ, Kolling FW, Affonso de Oliveira JF, Steinmetz NF, Rothstein JL, Lines L, Noelle RJ, Fiering S, Arias-Pulido H. An Abscopal Effect on Lung Metastases in Canine Mammary Cancer Patients Induced by Neoadjuvant Intratumoral Immunotherapy with Cowpea Mosaic Virus Nanoparticles and Anti-Canine PD-1. Cells 2024; 13:1478. [PMID: 39273048 PMCID: PMC11394642 DOI: 10.3390/cells13171478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/05/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Neoadjuvant intratumoral (IT) therapy could amplify the weak responses to checkpoint blockade therapy observed in breast cancer (BC). In this study, we administered neoadjuvant IT anti-canine PD-1 therapy (IT acPD-1) alone or combined with IT cowpea mosaic virus therapy (IT CPMV/acPD-1) to companion dogs diagnosed with canine mammary cancer (CMC), a spontaneous tumor resembling human BC. CMC patients treated weekly with acPD-1 (n = 3) or CPMV/acPD-1 (n = 3) for four weeks or with CPMV/acPD-1 (n = 3 patients not candidates for surgery) for up to 11 weeks did not experience immune-related adverse events. We found that acPD-1 and CPMV/acPD-1 injections resulted in tumor control and a reduction in injected tumors in all patients and in noninjected tumors located in the ipsilateral and contralateral mammary chains of treated dogs. In two metastatic CMC patients, CPMV/acPD-1 treatments resulted in the control and reduction of established lung metastases. CPMV/acPD-1 treatments were associated with altered gene expression related to TLR1-4 signaling and complement pathways. These novel therapies could be effective for CMC patients. Owing to the extensive similarities between CMC and human BC, IT CPMV combined with approved anti-PD-1 therapies could be a novel and effective immunotherapy to treat local BC and suppress metastatic BC.
Collapse
Affiliation(s)
- Petra Sergent
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA (L.L.); (R.J.N.); (S.F.)
| | | | | | - Daniel Luna Dávalos
- VETCONNECT Diagnóstico por imagen, Via Toledo, 2952 Mas Palomas, Monterrey 64780, Nuevo León, Mexico;
| | | | - Dora Alicia Mendoza Lechuga
- Centro Veterinario Valles, Zapopan 45070, Jalisco, Mexico; (A.J.A.C.); (M.D.G.P.); (D.A.M.L.); (A.J.C.); (D.C.M.)
| | - Daniel Alonso-Miguel
- Department of Animal Medicine and Surgery, Veterinary Medicine School, Complutense University of Madrid, 28040 Madrid, Spain;
| | | | - Abigail Jiménez Cuarenta
- Centro Veterinario Valles, Zapopan 45070, Jalisco, Mexico; (A.J.A.C.); (M.D.G.P.); (D.A.M.L.); (A.J.C.); (D.C.M.)
| | - Diana Cárdenas Muñoz
- Centro Veterinario Valles, Zapopan 45070, Jalisco, Mexico; (A.J.A.C.); (M.D.G.P.); (D.A.M.L.); (A.J.C.); (D.C.M.)
| | | | - Scott M. Palisoul
- Department of Pathology and Laboratory Medicine at Dartmouth Hitchcock Health, Center for Clinical Genomics and Advanced Technology, Lebanon, NH 03756, USA;
| | - Petra J. Lewis
- Department of Radiology Dartmouth Health Geisel School of Medicine, Lebanon, NH 03755, USA;
| | - Fred W. Kolling
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA;
| | - Jessica Fernanda Affonso de Oliveira
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA; (J.F.A.d.O.); (N.F.S.)
- Moores Cancer Center, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
- Center for Nano-ImmunoEngineering, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
- Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA
| | - Nicole F. Steinmetz
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA; (J.F.A.d.O.); (N.F.S.)
- Moores Cancer Center, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
- Center for Nano-ImmunoEngineering, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
- Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA
- Department of Radiology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
- Department of Bioengineering, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
- Institute for Materials Discovery and Design, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
- Center for Engineering in Cancer, University of California San Diego, La Jolla, CA 92093, USA
| | | | - Louise Lines
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA (L.L.); (R.J.N.); (S.F.)
| | - Randolph J. Noelle
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA (L.L.); (R.J.N.); (S.F.)
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA (L.L.); (R.J.N.); (S.F.)
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA;
| | - Hugo Arias-Pulido
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA (L.L.); (R.J.N.); (S.F.)
| |
Collapse
|
2
|
Rwandamuriye FX, Wang T, Zhang H, Elaskalani O, Kuster J, Ye X, Vitali B, Schreurs J, Orozco Morales ML, Norret M, Evans CW, Zemek RM, Iyer KS, Lesterhuis WJ, Wylie B. Local therapy with combination TLR agonists stimulates systemic anti-tumor immunity and sensitizes tumors to immune checkpoint blockade. Oncoimmunology 2024; 13:2395067. [PMID: 39188754 PMCID: PMC11346538 DOI: 10.1080/2162402x.2024.2395067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/26/2024] [Accepted: 08/16/2024] [Indexed: 08/28/2024] Open
Abstract
Toll-like receptor (TLR) agonists are being developed as anti-cancer therapeutics due to their potent immunostimulatory properties. However, clinical trials testing TLR agonists as monotherapy have often failed to demonstrate significant improvement over standard of care. We hypothesized that the anti-cancer efficacy of TLR agonist immunotherapy could be improved by combinatorial approaches. To prevent increased toxicity, often seen with systemic combination therapies, we developed a hydrogel to deliver TLR agonist combinations at low doses, locally, during cancer debulking surgery. Using tumor models of WEHI 164 and bilateral M3-9-M sarcoma and CT26 colon carcinoma, we assessed the efficacy of pairwise combinations of poly(I:C), R848, and CpG in controlling local and distant tumor growth. We show that combination of the TLR3 agonist poly(I:C) and TLR7/8 agonist R848 drives anti-tumor immunity against local and distant tumors. In addition, combination of local poly(I:C) and R848 sensitized tumors to systemic immune checkpoint blockade, improving tumor control. Mechanistically, we demonstrate that local therapy with poly(I:C) and R848 recruits inflammatory monocytes to the tumor draining lymph nodes early in the anti-tumor response. Finally, we provide proof of concept for intraoperative delivery of poly(I:C) and R848 together via a surgically applicable biodegradable hydrogel.
Collapse
Affiliation(s)
| | - Tao Wang
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Hanfu Zhang
- School of Molecular Sciences, University of Western Australia, Crawley, WA, Australia
| | - Omar Elaskalani
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Jorren Kuster
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Xueting Ye
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Breana Vitali
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Juliët Schreurs
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | | | - Marck Norret
- School of Molecular Sciences, University of Western Australia, Crawley, WA, Australia
| | - Cameron W. Evans
- School of Molecular Sciences, University of Western Australia, Crawley, WA, Australia
| | - Rachael M. Zemek
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - K. Swaminathan Iyer
- School of Molecular Sciences, University of Western Australia, Crawley, WA, Australia
| | - W. Joost Lesterhuis
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Ben Wylie
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
3
|
Mehta NK, Rakhra K, Meetze KA, Li B, Momin N, Chang JY, Wittrup KD, Baeuerle PA, Michaelson JS. CLN-617 Retains IL2 and IL12 in Injected Tumors to Drive Robust and Systemic Immune-Mediated Antitumor Activity. Cancer Immunol Res 2024; 12:1022-1038. [PMID: 38842347 PMCID: PMC11292205 DOI: 10.1158/2326-6066.cir-23-0636] [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: 08/03/2023] [Revised: 02/07/2024] [Accepted: 05/31/2024] [Indexed: 06/07/2024]
Abstract
Despite clinical evidence of antitumor activity, the development of cytokine therapies has been hampered by a narrow therapeutic window and limited response rates. Two cytokines of high interest for clinical development are interleukin 2 (IL2) and interleukin 12 (IL12), which potently synergize to promote the activation and proliferation of T cells and NK cells. However, the only approved human IL2 therapy, Proleukin, is rarely used in the clinic due to systemic toxicities, and no IL12 product has been approved to date due to severe dose-limiting toxicities. Here, we describe CLN-617, a first-in-class therapeutic for intratumoral (IT) injection that co-delivers IL2 and IL12 on a single molecule in a safe and effective manner. CLN-617 is a single-chain fusion protein comprised of IL2, leukocyte-associated immunoglobulin-like receptor 2 (LAIR2), human serum albumin (HSA), and IL12. LAIR2 and HSA function to retain CLN-617 in the treated tumor by binding collagen and increasing molecular weight, respectively. We found that IT administration of a murine surrogate of CLN-617, mCLN-617, eradicated established treated and untreated tumors in syngeneic models, significantly improved response to anti-PD1 checkpoint therapy, and generated a robust abscopal response dependent on cellular immunity and antigen cross-presentation. CLN-617 is being evaluated in a clinical trial in patients with advanced solid tumors (NCT06035744).
Collapse
Affiliation(s)
| | - Kavya Rakhra
- Cullinan Therapeutics, Inc., Cambridge, Massachusetts.
| | | | - Bochong Li
- Cullinan Therapeutics, Inc., Cambridge, Massachusetts.
| | - Noor Momin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania.
| | | | - K. Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| | - Patrick A. Baeuerle
- Cullinan Therapeutics, Inc., Cambridge, Massachusetts.
- Institute for Immunology, Ludwig Maximilians University, München, Germany.
| | | |
Collapse
|
4
|
Uher O, Hadrava Vanova K, Taïeb D, Calsina B, Robledo M, Clifton-Bligh R, Pacak K. The Immune Landscape of Pheochromocytoma and Paraganglioma: Current Advances and Perspectives. Endocr Rev 2024; 45:521-552. [PMID: 38377172 PMCID: PMC11244254 DOI: 10.1210/endrev/bnae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/19/2023] [Accepted: 02/02/2024] [Indexed: 02/22/2024]
Abstract
Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors derived from neural crest cells from adrenal medullary chromaffin tissues and extra-adrenal paraganglia, respectively. Although the current treatment for PPGLs is surgery, optimal treatment options for advanced and metastatic cases have been limited. Hence, understanding the role of the immune system in PPGL tumorigenesis can provide essential knowledge for the development of better therapeutic and tumor management strategies, especially for those with advanced and metastatic PPGLs. The first part of this review outlines the fundamental principles of the immune system and tumor microenvironment, and their role in cancer immunoediting, particularly emphasizing PPGLs. We focus on how the unique pathophysiology of PPGLs, such as their high molecular, biochemical, and imaging heterogeneity and production of several oncometabolites, creates a tumor-specific microenvironment and immunologically "cold" tumors. Thereafter, we discuss recently published studies related to the reclustering of PPGLs based on their immune signature. The second part of this review discusses future perspectives in PPGL management, including immunodiagnostic and promising immunotherapeutic approaches for converting "cold" tumors into immunologically active or "hot" tumors known for their better immunotherapy response and patient outcomes. Special emphasis is placed on potent immune-related imaging strategies and immune signatures that could be used for the reclassification, prognostication, and management of these tumors to improve patient care and prognosis. Furthermore, we introduce currently available immunotherapies and their possible combinations with other available therapies as an emerging treatment for PPGLs that targets hostile tumor environments.
Collapse
Affiliation(s)
- Ondrej Uher
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| | - Katerina Hadrava Vanova
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| | - David Taïeb
- Department of Nuclear Medicine, CHU de La Timone, Marseille 13005, France
| | - Bruna Calsina
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
- Familiar Cancer Clinical Unit, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institute of Health Carlos III (ISCIII), Madrid 28029, Spain
| | - Roderick Clifton-Bligh
- Department of Endocrinology, Royal North Shore Hospital, Sydney 2065, NSW, Australia
- Cancer Genetics Laboratory, Kolling Institute, University of Sydney, Sydney 2065, NSW, Australia
| | - Karel Pacak
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| |
Collapse
|
5
|
Sheth RA, Wehrenberg-Klee E, Patel SP, Brock KK, Fotiadis N, de Baère T. Intratumoral Injection of Immunotherapeutics: State of the Art and Future Directions. Radiology 2024; 312:e232654. [PMID: 39078294 PMCID: PMC11294769 DOI: 10.1148/radiol.232654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 02/17/2024] [Accepted: 02/28/2024] [Indexed: 07/31/2024]
Abstract
Systemic immunotherapies have led to tremendous progress across the cancer landscape. However, several challenges exist, potentially limiting their efficacy in the treatment of solid tumors. Direct intratumoral injection can increase the therapeutic index of immunotherapies while overcoming many of the barriers associated with systemic administration, including limited bioavailability to tumors and potential systemic safety concerns. However, challenges remain, including the lack of standardized approaches for administration, issues relating to effective drug delivery, logistical hurdles, and safety concerns specific to this mode of administration. This article reviews the biologic rationale for the localized injection of immunotherapeutic agents into tumors. It also addresses the existing limitations and practical considerations for safe and effective implementation and provide recommendations for optimizing logistics and treatment workflows. It also highlights the critical role that radiologists, interventional radiologists, and medical physicists play in intratumoral immunotherapy with respect to target selection, image-guided administration, and response assessment.
Collapse
Affiliation(s)
- Rahul A. Sheth
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Eric Wehrenberg-Klee
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Sapna P. Patel
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Kristy K. Brock
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Nicos Fotiadis
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Thierry de Baère
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| |
Collapse
|
6
|
Tijtgat J, Geeraerts X, Boisson A, Stevens L, Vounckx M, Dirven I, Schwarze JK, Raeymaeckers S, Forsyth R, Van Riet I, Tuyaerts S, Willard-Gallo K, Neyns B. Intratumoral administration of the immunologic adjuvant AS01 B in combination with autologous CD1c (BDCA-1) +/CD141 (BDCA-3) + myeloid dendritic cells plus ipilimumab and intravenous nivolumab in patients with refractory advanced melanoma. J Immunother Cancer 2024; 12:e008148. [PMID: 38212127 PMCID: PMC10806541 DOI: 10.1136/jitc-2023-008148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Patients with advanced melanoma who progress after treatment with immune checkpoint-inhibitors (ICI) and BRAF-/MEK-inhibitors (if BRAF V600 mutated) have no remaining effective treatment options. The presence of CD1c (BDCA-1)+ and CD141 (BDCA-3)+ myeloid dendritic cells (myDC) in the tumor microenvironment correlates with pre-existing immune recognition and responsiveness to immune checkpoint blockade. The synthetic saponin-based immune adjuvant AS01B enhances adaptive immunity through the involvement of myDC. METHODS In this first-in-human phase I clinical trial, patients with metastatic melanoma refractory to ICI and BRAF-/MEK inhibitors (when indicated) were recruited. Patients received an intravenous administration of low-dose nivolumab (10 mg, every 2 weeks) plus an intratumoral (IT) administration of 10 mg ipilimumab and 50 µg (0.5 mL) AS01B (every 2 weeks). All myDC, isolated from blood, were injected on day 2 into the same metastatic lesion. Tumor biopsies and blood samples were collected at baseline and repeatedly on treatment. Multiplex immunohistochemistry (mIHC) was performed on biopsy sections to characterize and quantify the IT and peritumoral immune cell composition. RESULTS Study treatment was feasible and well tolerated without the occurrence of unexpected adverse events in all eight patients. Four patients (50%) obtained a complete response (CR) in the injected lesions. Of these, two patients obtained an overall CR, and one patient a partial response. All responses are ongoing after more than 1 year of follow-up. One additional patient had a stable disease as best response. The disease control rate was 50%. Median progression-free survival and overall survival were 24.1 and 41.9 weeks, respectively. Baseline tumor biopsies from patients who responded to treatment had features of T-cell exclusion. During treatment, there was an increased T-cell infiltration, with a reduced mean distance between T cells and tumor cells. Peripheral blood immune cell composition did not significantly change during study treatment. CONCLUSIONS Combining an intratumoral injection of CD1c (BDCA-1)+ and CD141 (BDCA-3)+ myDC with repeated IT administration of ipilimumab and AS01B and systemic low-dose nivolumab is safe, feasible with promising early results, worthy of further clinical investigation. TRIAL REGISTRATION NUMBER ClinicalTrials.gov identifier NCT03707808.
Collapse
Affiliation(s)
- Jens Tijtgat
- Department of Medical Oncology/Laboratory for Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Xenia Geeraerts
- Department of Medical Oncology/Laboratory for Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Anais Boisson
- Molecular Immunology Unit (MIU), Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Latoya Stevens
- Department of Medical Oncology/Laboratory for Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Manon Vounckx
- Department of Medical Oncology/Laboratory for Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Iris Dirven
- Department of Medical Oncology/Laboratory for Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Julia Katharina Schwarze
- Department of Medical Oncology/Laboratory for Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Steven Raeymaeckers
- Department of Radiology, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Ramses Forsyth
- Department of Pathology, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Ivan Van Riet
- Department of Hematology, Stem Cell Laboratory, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Sandra Tuyaerts
- Department of Medical Oncology/Laboratory for Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Karen Willard-Gallo
- Molecular Immunology Unit (MIU), Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Bart Neyns
- Department of Medical Oncology/Laboratory for Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| |
Collapse
|
7
|
Awad RM, De Vlaeminck Y, Meeus F, Ertveldt T, Zeven K, Ceuppens H, Goyvaerts C, Verdonck M, Salguero G, Raes G, Devoogdt N, Breckpot K. In vitro modelling of local gene therapy with IL-15/IL-15Rα and a PD-L1 antagonist in melanoma reveals an interplay between NK cells and CD4 + T cells. Sci Rep 2023; 13:18995. [PMID: 37923822 PMCID: PMC10624833 DOI: 10.1038/s41598-023-45948-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023] Open
Abstract
Blockade of the immune checkpoint axis consisting of programmed death-1 (PD-1) and its ligand PD-L1 alleviates the functional inhibition of tumor-infiltrating lymphoid cells yet weakly induces their expansion. Exogenous cytokines could further expand lymphoid cells and thus synergize with αPD-L1 therapy. However, systemic delivery of most cytokines causes severe toxicity due to unspecific expansion of immune cells in the periphery. Here, we modelled local delivery of cytokines and αPD-L1 therapeutics to immune cell-containing in vitro melanoma tumors. Three-dimensional tumor models consisting of 624-MEL cells were co-cultured with human peripheral blood lymphoid cells (PBLs) in presence of the cytokines IL-2, IL-7, IL-15, IL-21 and IFN-γ. To model local gene therapy, melanoma tumors were modified with lentiviral vectors encoding IL-15 fused to IL-15Rα (IL-15/IL-15Rα) and K2-Fc, a fusion of a human PD-L1 specific single domain antibody to immunoglobulin (Ig)G1 Fc. To evaluate the interplay between PBL fractions, NK cells, CD4+ T cells or CD8+ T cells were depleted. Tumor cell killing was followed up using real time imaging and immune cell expansion and activation was evaluated with flow cytometry. Among the tested cytokines, IL-15 was the most potent cytokine in stimulating tumor cell killing and expanding both natural killer (NK) cells and CD8+ T cells. Gene-based delivery of IL-15/IL-15Rα to tumor cells, shows expansion of NK cells, activation of NK cells, CD4+ and CD8+ T cells, and killing of tumor spheroids. Both NK cells and CD8+ T cells are necessary for tumor cell killing and CD4+ T-cell activation was reduced without NK cells. Co-delivery of K2-Fc improved tumor cell killing coinciding with increased activation of NK cells, which was independent of bystander T cells. CD4+ or CD8+ T cells were not affected by the co-delivery of K2-Fc even though NK-cell activation impacted CD4+ T-cell activation. This study demonstrates that gene-based delivery of IL-15/IL-15Rα to tumor cells effectively mediates anti-tumor activity and sensitizes the tumor microenvironment for therapy with αPD-L1 therapeutics mainly by impacting NK cells. These findings warrant further investigation of gene-based IL-15 and K2-Fc delivery in vivo.
Collapse
Affiliation(s)
- Robin Maximilian Awad
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium.
| | - Yannick De Vlaeminck
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Fien Meeus
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Thomas Ertveldt
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Katty Zeven
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Hannelore Ceuppens
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Cleo Goyvaerts
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Magali Verdonck
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Gustavo Salguero
- Advanced Therapies Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud-IDCBIS, 111611, Bogotá, Colombia
| | - Geert Raes
- Laboratory of Cellular and Molecular Immunology, Department of Bioengineering Sciences, Vrije Universiteit Brussel, 1050, Brussels, Belgium
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050, Brussels, Belgium
- Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, 1050, Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Karine Breckpot
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium.
| |
Collapse
|
8
|
Ghosn M, Tselikas L, Champiat S, Deschamps F, Bonnet B, Carre É, Testan M, Danlos FX, Farhane S, Susini S, Suzzoni S, Ammari S, Marabelle A, De Baere T. Intratumoral Immunotherapy: Is It Ready for Prime Time? Curr Oncol Rep 2023; 25:857-867. [PMID: 37129706 DOI: 10.1007/s11912-023-01422-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2023] [Indexed: 05/03/2023]
Abstract
PURPOSE OF REVIEW This review presents the rationale for intratumoral immunotherapy, technical considerations and safety. Clinical results from the latest trials are provided and discussed. RECENT FINDINGS Intratumoral immunotherapy is feasible and safe in a wide range of cancer histologies and locations, including lung and liver. Studies mainly focused on multi-metastatic patients, with some positive trials such as T-VEC in melanoma, but evidence of clinical benefit is still lacking. Recent results showed improved outcomes in patients with a low tumor burden. Intratumoral immunotherapy can lower systemic toxicities and boost local and systemic immune responses. Several studies have proven the feasibility, repeatability, and safety of this approach, with some promising results in clinical trials. The clinical benefit might be improved in patients with a low tumor burden. Future clinical trials should focus on adequate timing of treatment delivery during the course of the disease, particularly in the neoadjuvant setting.
Collapse
Affiliation(s)
- Mario Ghosn
- Radiologie Interventionnelle, Département d'Anesthésie Chirurgie Et Imagerie Interventionnelle (DACI), Gustave Roussy, Villejuif, 94800, France
- Centre D'Investigation Clinique BIOTHERIS, INSERM CIC1428, Villejuif, France
| | - Lambros Tselikas
- Radiologie Interventionnelle, Département d'Anesthésie Chirurgie Et Imagerie Interventionnelle (DACI), Gustave Roussy, Villejuif, 94800, France.
- Centre D'Investigation Clinique BIOTHERIS, INSERM CIC1428, Villejuif, France.
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Villejuif, France.
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, France.
| | - Stéphane Champiat
- Centre D'Investigation Clinique BIOTHERIS, INSERM CIC1428, Villejuif, France
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Villejuif, France
- Département D'Innovation Thérapeutique Et D'Essais Précoces (DITEP), Gustave Roussy, Villejuif, France
| | - Frederic Deschamps
- Radiologie Interventionnelle, Département d'Anesthésie Chirurgie Et Imagerie Interventionnelle (DACI), Gustave Roussy, Villejuif, 94800, France
| | - Baptiste Bonnet
- Radiologie Interventionnelle, Département d'Anesthésie Chirurgie Et Imagerie Interventionnelle (DACI), Gustave Roussy, Villejuif, 94800, France
| | - Émilie Carre
- Centre D'Investigation Clinique BIOTHERIS, INSERM CIC1428, Villejuif, France
| | - Marine Testan
- Centre D'Investigation Clinique BIOTHERIS, INSERM CIC1428, Villejuif, France
| | - François-Xavier Danlos
- Centre D'Investigation Clinique BIOTHERIS, INSERM CIC1428, Villejuif, France
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Villejuif, France
- Département D'Innovation Thérapeutique Et D'Essais Précoces (DITEP), Gustave Roussy, Villejuif, France
| | - Siham Farhane
- Centre D'Investigation Clinique BIOTHERIS, INSERM CIC1428, Villejuif, France
| | - Sandrine Susini
- Centre D'Investigation Clinique BIOTHERIS, INSERM CIC1428, Villejuif, France
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Villejuif, France
| | - Steve Suzzoni
- Département Pharmacie, Gustave Roussy, Villejuif, France
| | - Samy Ammari
- Department of Imaging, Gustave Roussy, Université Paris Saclay, 94805, Villejuif, France
- Biomaps, UMR1281 INSERM, CEA, CNRS, Université Paris-Saclay, 94805, Villejuif, France
| | - Aurélien Marabelle
- Centre D'Investigation Clinique BIOTHERIS, INSERM CIC1428, Villejuif, France
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Villejuif, France
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, France
- Département D'Innovation Thérapeutique Et D'Essais Précoces (DITEP), Gustave Roussy, Villejuif, France
| | - Thierry De Baere
- Radiologie Interventionnelle, Département d'Anesthésie Chirurgie Et Imagerie Interventionnelle (DACI), Gustave Roussy, Villejuif, 94800, France
- Centre D'Investigation Clinique BIOTHERIS, INSERM CIC1428, Villejuif, France
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
9
|
Meng L, Wei Y, Xiao Y. Chemo-immunoablation of solid tumors: A new concept in tumor ablation. Front Immunol 2023; 13:1057535. [PMID: 36713427 PMCID: PMC9878389 DOI: 10.3389/fimmu.2022.1057535] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/09/2022] [Indexed: 01/13/2023] Open
Abstract
Chemical ablation was designed to inject chemical agents directly into solid tumors to kill cells and is currently only used clinically for the palliative treatment of tumors. The application and combination of different drugs, from anhydrous ethanol, and glacial acetic acid to epi-amycin, have been clinically tested for a long time. The effectiveness is unsatisfactory due to chemical agents' poor diffusion and concentration. Immunotherapy is considered a prospective oncologic therapeutic. Still, the clinical applications were limited by the low response rate of patients to immune drugs and the immune-related adverse effects caused by high doses. The advent of intratumoral immunotherapy has well addressed these issues. However, the efficacy of intratumoral immunotherapy alone is uncertain, as suggested by the results of preclinical and clinical studies. In this study, we will focus on the research of immunosuppressive tumor microenvironment with chemoablation and intratumoral immunotherapy, the synergistic effect between chemotherapeutic drugs and immunotherapy. We propose a new concept of intratumoral chemo-immunoablation. The concept opens a new perspective for tumor treatment from direct killing of tumor cells while, enhancing systemic anti-tumor immune response, and significantly reducing adverse effects of drugs.
Collapse
Affiliation(s)
- Liangliang Meng
- Department of Radiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China,Department of Radiology, Chinese PAP Hospital of Beijing, Beijing, China
| | - Yingtian Wei
- Department of Radiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yueyong Xiao
- Department of Radiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China,*Correspondence: Yueyong Xiao,
| |
Collapse
|
10
|
Abstract
Historically, cancer research and therapy have focused on malignant cells and their tumor microenvironment. However, the vascular, lymphatic and nervous systems establish long-range communication between the tumor and the host. This communication is mediated by metabolites generated by the host or the gut microbiota, as well by systemic neuroendocrine, pro-inflammatory and immune circuitries-all of which dictate the trajectory of malignant disease through molecularly defined biological mechanisms. Moreover, aging, co-morbidities and co-medications have a major impact on the development, progression and therapeutic response of patients with cancer. In this Perspective, we advocate for a whole-body 'ecological' exploration of malignant disease. We surmise that accumulating knowledge on the intricate relationship between the host and the tumor will shape rational strategies for systemic, bodywide interventions that will eventually improve tumor control, as well as quality of life, in patients with cancer.
Collapse
|
11
|
Bezu L, Kepp O, Kroemer G. Local anesthetics and immunotherapy: a novel combination to fight cancer. Semin Immunopathol 2022; 45:265-272. [PMID: 36044068 DOI: 10.1007/s00281-022-00960-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/17/2022] [Indexed: 11/29/2022]
Abstract
Intratumoral injection of oncolytic agents such as modified herpes simplex virus T-VEC or local administration of non-viral oncolytic therapies (such as radiofrequency, chemoembolization, cryoablation, or radiotherapy) can activate an anticancer immune response and hence trigger abscopal effects reducing secondary lesions. Preliminary data suggested that oncolytic treatments modulate tumor-infiltrating immune effectors and can be advantageously combined with the immune checkpoint inhibitors. Recent findings indicate that local anesthetics, which are usually used in the clinics to control surgical pain, also possess antineoplastic effects mimicking oncolytic treatments if they are injected into malignant lesions. Moreover, the association of local anesthetics with systemic immune checkpoint inhibition significantly improved overall survival in several preclinical tumor models. This may be explained by direct cytotoxic activity of local anesthetics and additional immune-related abscopal effects. We also summarize the molecular and cellular mechanisms by which the combination of local anesthetics and immunotherapy improves tumor control by the immune system.
Collapse
Affiliation(s)
- Lucillia Bezu
- UMR1138, Centre de Recherche Des Cordeliers, Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, INSERM, Institut Universitaire de France, Paris, France. .,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France. .,Service d'Anesthésie Gustave Roussy Cancer Campus, Villejuif, France. .,EuroPeriscope: Onco-Anaesthesiology Research Group, Brussels, Belgium.
| | - Oliver Kepp
- UMR1138, Centre de Recherche Des Cordeliers, Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, INSERM, Institut Universitaire de France, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France
| | - Guido Kroemer
- UMR1138, Centre de Recherche Des Cordeliers, Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, INSERM, Institut Universitaire de France, Paris, France. .,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France. .,Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|