1
|
Revising the Landscape of Cytokine-Induced Killer Cell Therapy in Lung Cancer: Focus on Immune Checkpoint Inhibitors. Int J Mol Sci 2023; 24:ijms24065626. [PMID: 36982701 PMCID: PMC10054817 DOI: 10.3390/ijms24065626] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/11/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Undeniably, immunotherapy has markedly improved the survival rate of cancer patients. The scenario is no different in lung cancer, where multiple treatment options are now available and the inclusion of immunotherapy yields better clinical benefits than previously used chemotherapeutic strategies. Of interest, cytokine-induced killer (CIK) cell immunotherapy has also taken a central role in clinical trials for the treatment of lung cancer. Herein, we describe the relative success of CIK cell therapy (alone and combined with dendritic cells as DC/CIKs) in lung cancer clinical trials and discuss its combination with known immune checkpoint inhibitors (anti-CTLA-4 and anti-PD-1/PD-L1). Additionally, we provide insights into the findings of several preclinical in vitro/in vivo studies linked to lung cancer. In our opinion, CIK cell therapy, which recently completed 30 years and has been approved in many countries, including Germany, offers tremendous potential for lung cancer. Foremost, when it is optimized on a patient-by-patient basis with special attention to the patient-specific genomic signature.
Collapse
|
2
|
Luginbuehl V, Abraham E, Kovar K, Flaaten R, Müller AMS. Better by design: What to expect from novel CAR-engineered cell therapies? Biotechnol Adv 2022; 58:107917. [PMID: 35149146 DOI: 10.1016/j.biotechadv.2022.107917] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 12/15/2022]
Abstract
Chimeric antigen receptor (CAR) technology, and CAR-T cells in particular, have emerged as a new and powerful tool in cancer immunotherapy since demonstrating efficacy against several hematological malignancies. However, despite encouraging clinical results of CAR-T cell therapy products, a significant proportion of patients do not achieve satisfactory responses, or relapse. In addition, CAR-T cell applications to solid tumors is still limited due to the tumor microenvironment and lack of specifically targetable tumor antigens. All current products on the market, as well as most investigational CAR-T cell therapies, are autologous, using the patient's own peripheral blood mononuclear cells as starting material to manufacture a patient-specific batch. Alternative cell sources are, therefore, under investigation (e.g. allogeneic cells from an at least partially human leukocyte antigen (HLA)-matched healthy donor, universal "third-party" cells from a non-HLA-matched donor, cord blood-derived cells, immortalized cell lines or cells differentiated from induced pluripotent stem cells). However, genetic modifications of CAR-engineered cells, bioprocesses used to expand cells, and improved supply chains are still complex and costly. To overcome drawbacks associated with CAR-T technologies, novel CAR designs have been used to genetically engineer cells derived from alpha beta (αβ) T cells, other immune cells such as natural killer (NK) cells, gamma delta (γδ) T cells, macrophages or dendritic cells. This review endeavours to trigger ideas on the next generation of CAR-engineered cell therapies beyond CAR-T cells and, thus, will enable effective, safe and affordable therapies for clinical management of cancer. To achieve this, we present a multidisciplinary overview, addressing a wide range of critical aspects: CAR design, development and manufacturing technologies, pharmacological concepts and clinical applications of CAR-engineered cell therapies. Each of these fields employs a large number of ground-breaking scientific advances, where coordinated and complex process and product development occur at their interfaces.
Collapse
Affiliation(s)
- Vera Luginbuehl
- Novartis Oncology, Cell & Gene Therapy, Novartis Pharma Schweiz AG, Rotkreuz, Switzerland.
| | - Eytan Abraham
- Personalized Medicine Lonza Pharma&Biotech, Lonza Ltd., Walkersville, MD, USA
| | | | - Richard Flaaten
- Novartis Oncology, Cell & Gene Therapy, Novartis Norge AS, Oslo, Norway
| | - Antonia M S Müller
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Dalla Pietà A, Cappuzzello E, Palmerini P, Ventura A, Visentin A, Astori G, Chieregato K, Mozzo V, Perbellini O, Tisi MC, Trentin L, Visco C, Ruggeri M, Sommaggio R, Rosato A. Innovative therapeutic strategy for B-cell malignancies that combines obinutuzumab and cytokine-induced killer cells. J Immunother Cancer 2021; 9:jitc-2021-002475. [PMID: 34272306 PMCID: PMC8287629 DOI: 10.1136/jitc-2021-002475] [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] [Accepted: 06/02/2021] [Indexed: 12/24/2022] Open
Abstract
Background Patients affected by aggressive B-cell malignancies who are resistant to primary or salvage chemoimmunotherapy have an extremely poor prognosis and limited therapeutic options. Promising therapeutic success has been achieved with the infusion of CD19 chimeric antigen receptor-T cells, but several limits still restrain the administration to a limited proportion of patients. This unmet clinical need might be fulfilled by an adoptive immunotherapy approach that combines cytokine-induced killer (CIK) cells and monoclonal antibodies (mAb) to the CD20 antigen. Indeed, CIK cells are an effector population endowed with antitumor activity, which can be further improved and antigen-specifically redirected by clinical-grade mAb triggering antibody-dependent cell-mediated cytotoxicity. Methods CIK cells were generated from peripheral blood of patients affected by different B-cell malignancies using a blinatumomab-based cell culture protocol. Effector cells were combined with the anti-CD20 mAb obinutuzumab and their therapeutic activity was assessed both in vitro and in vivo. Results CIK cells were successfully expanded in clinically relevant numbers, starting from small volumes of peripheral blood with extremely low CD3+ counts and high tumor burden. This relied on the addition of blinatumumab in culture, which leads to the simultaneous expansion of effector cells and the complete elimination of the neoplastic component. Moreover, CIK cells were highly cytotoxic in vitro against both B-cell tumor cell lines and autologous neoplastic targets, and had a significant therapeutic efficacy against a B-cell malignancy patient-derived xenograft on in vivo transfer. Conclusions The combination of an easily expandable CIK cell effector population with a mAb already in clinical use establishes a tumor antigen-specific redirection strategy that can be rapidly translated into clinical practice, providing an effective therapeutic alternative for B-cell malignancies without any need for genetic modifications. Additionally, the approach can be potentially applied to an extremely vast array of different tumors by simply substituting the targeting mAb.
Collapse
Affiliation(s)
- Anna Dalla Pietà
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padova, Italy
| | - Elisa Cappuzzello
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padova, Italy
| | - Pierangela Palmerini
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padova, Italy
| | - Annavera Ventura
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padova, Italy
| | - Andrea Visentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padova, Italy
| | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Department of Hematology, San Bortolo Hospital of Vicenza, Vicenza, Italy
| | - Katia Chieregato
- Advanced Cellular Therapy Laboratory, Department of Hematology, San Bortolo Hospital of Vicenza, Vicenza, Italy.,Consorzio per la Ricerca Sanitaria (CORIS) of Veneto Region, Padova, Italy
| | | | - Omar Perbellini
- Cell Therapy and Hematology, San Bortolo Hospital, Vicenza, Italy
| | | | - Livio Trentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padova, Italy
| | - Carlo Visco
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Marco Ruggeri
- Cell Therapy and Hematology, San Bortolo Hospital, Vicenza, Italy
| | | | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padova, Italy .,Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| |
Collapse
|
4
|
Recruitment, Infiltration, and Cytotoxicity of HLA-Independent Killer Lymphocytes in Three-Dimensional Melanoma Models. Cancers (Basel) 2021; 13:cancers13102302. [PMID: 34065007 PMCID: PMC8151151 DOI: 10.3390/cancers13102302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 04/28/2021] [Accepted: 05/05/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Limited therapeutic results of immune checkpoint inhibitors in definite tumor settings, such as melanoma, call for alternative or complementary approaches. Among these, adoptive cell therapy (ACT) by means of HLA-independent tumor killer lymphocytes is a promising approach. We aimed at developing a pre-clinical 3D model to investigate and visualize the interaction between tumor and immune effectors in melanoma. To this aim, we employed Cytokine-Induced Killer cells (CIK) and NK-92 on patient-derived melanoma samples. By means of imaging-based methods, we measured the effector recruitment on the 3D targets, their infiltration, and cytotoxic activity. Our results and methodologies can be easily generalized to other effectors and other classes of tumors and help elucidate fundamental questions on the basic biology and kinetics of immune effector recruitment in a realistic 3D setting mimicking a solid tumor. Abstract Cancer adoptive cell therapy (ACT) with HLA-independent tumor killer lymphocytes is a promising approach, with intrinsic features potentially addressing crucial tumor-escape mechanisms of checkpoint inhibitors. Cytokine-induced Killer (CIK) and Natural Killer (NK) lymphocytes share similar tumor-killing mechanisms, with preclinical evidence of intense activity against multiple solid tumors and currently testing in clinical studies. To improve the effective clinical translation of such ACT approaches, several fundamental questions still need to be addressed within appropriate preclinical contexts, capable of overcoming limitations imposed by most traditional two-dimensional assays. Here, we developed a novel experimental approach to explore, dissect, and visualize the interactions of CIK and NK lymphocytes with melanoma tumors in vitro in 3D. Primary melanoma cells were assembled into small tumors that were dispersed in a 3D matrix and challenged with patient-derived CIK or the NK-92 cell line. By means of imaging-based methods, we reported, visualized, and quantitatively measured the recruitment of CIK and NK on the 3D targets, their infiltration, and cytotoxic activity. Our results support the effective tumor recruitment and tumor infiltration by CIK and NK. Such features appeared dependent on the specific geometric aspects of the environment but can be explained in terms of directional migration toward the tumor, without invoking major feedback components. Overall, our 3D platform allows us to monitor the processes of tumor recruitment, infiltration, and killing by means of live measurements, revealing important kinetic aspects of ACT with CIK and NK against melanoma.
Collapse
|
5
|
Leuci V, Donini C, Grignani G, Rotolo R, Mesiano G, Fiorino E, Gammaitoni L, D'Ambrosio L, Merlini A, Landoni E, Medico E, Capellero S, Giraudo L, Cattaneo G, Iaia I, Pignochino Y, Basiricò M, Vigna E, Pisacane A, Fagioli F, Ferrone S, Aglietta M, Dotti G, Sangiolo D. CSPG4-Specific CAR.CIK Lymphocytes as a Novel Therapy for the Treatment of Multiple Soft-Tissue Sarcoma Histotypes. Clin Cancer Res 2020; 26:6321-6334. [PMID: 32900797 DOI: 10.1158/1078-0432.ccr-20-0357] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 07/14/2020] [Accepted: 08/25/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE No effective therapy is available for unresectable soft-tissue sarcomas (STS). This unmet clinical need prompted us to test whether chondroitin sulfate proteoglycan 4 (CSPG4)-specific chimeric antigen receptor (CAR)-redirected cytokine-induced killer lymphocytes (CAR.CIK) are effective in eliminating tumor cells derived from multiple STS histotypes in vitro and in immunodeficient mice. EXPERIMENTAL DESIGN The experimental platform included patient-derived CAR.CIK and cell lines established from multiple STS histotypes. CAR.CIK were transduced with a retroviral vector encoding second-generation CSPG4-specific CAR (CSPG4-CAR) with 4-1BB costimulation. The functional activity of CSPG4-CAR.CIK was explored in vitro, in two- and three-dimensional STS cultures, and in three in vivo STS xenograft models. RESULTS CSPG4-CAR.CIK were efficiently generated from patients with STS. CSPG4 was highly expressed in multiple STS histotypes by in silico analysis and on all 16 STS cell lines tested by flow cytometry. CSPG4-CAR.CIK displayed superior in vitro cytolytic activity against multiple STS histotypes as compared with paired unmodified control CIK. CSPG4-CAR.CIK also showed strong antitumor activity against STS spheroids; this effect was associated with tumor recruitment, infiltration, and matrix penetration. CSPG4-CAR.CIK significantly delayed or reversed tumor growth in vivo in three STS xenograft models (leiomyosarcoma, undifferentiated pleomorphic sarcoma, and fibrosarcoma). Tumor growth inhibition persisted for up to 2 weeks following the last administration of CSPG4-CAR.CIK. CONCLUSIONS This study has shown that CSPG4-CAR.CIK effectively targets multiple STS histotypes in vitro and in immunodeficient mice. These results provide a strong rationale to translate the novel strategy we have developed into a clinical setting.
Collapse
Affiliation(s)
- Valeria Leuci
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Chiara Donini
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | | | - Ramona Rotolo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Giulia Mesiano
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Erika Fiorino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | | | | | - Alessandra Merlini
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Elisa Landoni
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Enzo Medico
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Sonia Capellero
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Lidia Giraudo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Giulia Cattaneo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Ilenia Iaia
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Ymera Pignochino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Marco Basiricò
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Elisa Vigna
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | | | - Franca Fagioli
- Pediatric Onco-Hematology, Division of Stem Cell Transplantation and Cellular Therapy, Regina Margherita Children's Hospital, University of Turin, Turin, Italy
| | - Soldano Ferrone
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Massimo Aglietta
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina
| | - Dario Sangiolo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy. .,Department of Oncology, University of Torino, Turin, Italy
| |
Collapse
|
6
|
Liu H, Xu Y, Liang K, Liu R. Immune Cells Combined With NLRP3 Inflammasome Inhibitor Exert Better Antitumor Effect on Pancreatic Ductal Adenocarcinoma. Front Oncol 2020; 10:1378. [PMID: 32974137 PMCID: PMC7471723 DOI: 10.3389/fonc.2020.01378] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/30/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is among the most aggressive malignancies associated with chronic inflammation. Moreover, cellular immunity can be inhibited by inflammation induced by nucleotide-binding domain, leucine-rich family, pyrin-containing 3 (NLRP3) inflammasome. Accordingly, NLRP3 inhibition combining cytokine-induced killer (CIK) cells may improve antitumor efficacy. 3,4-Methylenedioxy-β-nitrostyrene (MNS) was selected as a specific NLRP3 inflammasome inhibitor. Western blot was used to evaluate the NLRP3 inflammasome expression in pancreatic cancer cell lines SW1990 and PANC-1. The impact of NLRP3 inhibition on migration, invasiveness, and proliferation of pancreatic cancer cells was analyzed through wound healing assay, Transwell assay, and Cell Counting Kit-8 (CCK-8) assay, respectively. The combining antitumor effect in vivo of CIK and NLRP3 inhibition was evaluated in a subcutaneous human pancreatic cancer BALB/c nude mouse model. Western blot analysis showed significant expression of NLRP3 inflammasome in human pancreatic cancer lines SW1990 and PANC-1, and MNS did significantly inhibit the expression of NLRP3 inflammasome in cell lines. Moreover, NLRP3 inhibition could significantly decrease the migration, invasiveness, and proliferation of pancreatic cancer cells. In vivo experiments showed that combination treatment with MNS and CIK cells had the greatest antitumor effect among the four treatment groups including control, MNS, and CIK. Combination treatment with NLRP3 inflammasome inhibition and CIK cells showed greater antitumor efficacy through inflammation inhibition and immunity restoration.
Collapse
Affiliation(s)
- Hailiang Liu
- Department of Burn and Plastic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yong Xu
- The Second Hepatobiliary Surgical Department, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Kai Liang
- General Surgery Institute, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Rong Liu
- The Second Hepatobiliary Surgical Department, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
7
|
Palmerini P, Dalla Pietà A, Sommaggio R, Ventura A, Astori G, Chieregato K, Tisi MC, Visco C, Perbellini O, Ruggeri M, Cappuzzello E, Rosato A. A serum-free protocol for the ex vivo expansion of Cytokine-Induced Killer cells using gas-permeable static culture flasks. Cytotherapy 2020; 22:511-518. [PMID: 32631696 DOI: 10.1016/j.jcyt.2020.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/13/2020] [Accepted: 05/06/2020] [Indexed: 12/21/2022]
Abstract
Cytokine-Induced (CIK) cells represent an attractive approach for cell-based immunotherapy, as they show several advantages compared with other strategies. Here we describe an original serum-free protocol for CIK cell expansion that employs G-Rex devices and compare the resulting growth, viability, phenotypic profile and cytotoxic activity with conventional culture in tissue flasks. CIK cells were obtained from buffy coats, seeded in parallel in G-Rex and tissue flasks, and stimulated with clinical-grade IFN-γ, anti-CD3 antibody and IL-2. G-Rex led to large numbers of CIK cells, with a minimal need for technical interventions, thus reducing the time and costs of culture manipulation. CIK cells generated in G-Rex showed a less differentiated phenotype, with a significantly higher expression of naive-associated markers such as CD62L, CD45RA and CCR7, which correlates with a remarkable expansion potential in culture and could lead to longer persistence and a more sustained anti-tumor response in vivo. The described procedure can be easily translated to large-scale production under Good Manufacturing Practice. Overall, this protocol has strong advantages over existing procedures, as it allows easier, time-saving and cost-effective production of CIK effector cells, fostering their clinical application.
Collapse
Affiliation(s)
- Pierangela Palmerini
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy
| | - Anna Dalla Pietà
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy
| | | | - Annavera Ventura
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy
| | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Department of Hematology, Vicenza Hospital, Vicenza, Italy
| | - Katia Chieregato
- Advanced Cellular Therapy Laboratory, Department of Hematology, Vicenza Hospital, Vicenza, Italy
| | | | - Carlo Visco
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | | | - Marco Ruggeri
- Hematology Department, San Bortolo Hospital, Vicenza, Italy
| | - Elisa Cappuzzello
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy; Veneto Institute of Oncology IOV - IRCCS, Padua, Italy.
| |
Collapse
|
8
|
Zhou M, Wang J, Li CP, Xu JY, Chen B. Autologous Cytokine-Induced Killer Cell Immunotherapy for Patients with High-Risk Diffuse Large B Cell Lymphoma After the First Complete Remission. Onco Targets Ther 2020; 13:5879-5885. [PMID: 32606798 PMCID: PMC7319512 DOI: 10.2147/ott.s254291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/26/2020] [Indexed: 12/30/2022] Open
Abstract
Purpose To evaluate whether autologous cytokine-induced killer (CIK) cell immunotherapy improves the prognosis of patients with high-risk diffuse large B cell lymphoma (DLBCL) after the first complete remission (CR). Patients and Methods Peripheral blood mononuclear cells (PBMCs) were extracted from 20 patients with high-risk DLBCL (IPI≥3) after the first CR. Twenty CR patients who were age- and sex-matched during the same period were selected as controls. PBMCs were cultured with IFN-γ, IL-2 and anti-CD3 mAb to generate CIK cells. These obtained cells were then transfused back into the patients; the transfusion was repeated every 3 months up to a total of four courses. Changes in peripheral blood lymphocyte subgroups and survival were assessed. Results Compared with the baseline proportions, the proportion of CD3+ T cells, CD3+CD8+ T cells, and NK cells in the peripheral blood were significantly higher after transfusions (p<0.05). The 5-year DFS was improved from 45.0 ± 11.1% to 79.3 ± 9.2% in the CIK group (HR favoring CIK, 0.29; 95% CI, 0.09 to 0.92; p = 0.035), and the 5-year OS was estimated at 90 ± 6.7% for CIK versus 55 ± 11.1% for control (HR favoring CIK, 0.20; 95% CI, 0.04 to 0.93; p = 0.040). No severe side effects were observed related to CIK treatment. Conclusion Autologous CIK cell immunotherapy has emerged as a safe and efficacious option to improve the prognosis of patients with high-risk DLBCL after the first CR.
Collapse
Affiliation(s)
- Min Zhou
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Jing Wang
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Cui-Ping Li
- Department of Transfusion, BenQ Medical Center, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Jing-Yan Xu
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Bing Chen
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
9
|
Sommaggio R, Cappuzzello E, Dalla Pietà A, Tosi A, Palmerini P, Carpanese D, Nicolè L, Rosato A. Adoptive cell therapy of triple negative breast cancer with redirected cytokine-induced killer cells. Oncoimmunology 2020; 9:1777046. [PMID: 32923140 PMCID: PMC7458615 DOI: 10.1080/2162402x.2020.1777046] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cytokine-Induced Killer (CIK) cells share several functional and phenotypical properties of both T and natural killer (NK) cells. They represent an attractive approach for cell-based immunotherapy, as they do not require antigen-specific priming for tumor cell recognition, and can be rapidly expanded in vitro. Their relevant expression of FcγRIIIa (CD16a) can be exploited in combination with clinical-grade monoclonal antibodies (mAbs) to redirect their lytic activity in an antigen-specific manner. Here, we report the efficacy of this combined approach against triple negative breast cancer (TNBC), an aggressive tumor that still requires therapeutic options. Different primitive and metastatic TNBC cancer mouse models were established in NSG mice, either by implanting patient-derived TNBC samples or injecting MDA-MB-231 cells orthotopically or intravenously. The combined treatment consisted in the repeated intratumoral or intravenous injection of CIK cells and cetuximab. Tumor growth and metastasis were monitored by bioluminescence or immunohistochemistry, and survival was recorded. CIK cells plus cetuximab significantly restrained primitive tumor growth in mice, either in patient-derived tumor xenografts or MDA-MB-231 cell line models. Moreover, this approach almost completely abolished metastasis spreading and dramatically improved survival. The antigen-specific mAb favored tumor and metastasis tissue infiltration by CIK cells, and led to an enrichment of the CD16a+ subset. Data highlight the potentiality of this novel immunotherapy strategy where a nonspecific cytotoxic cell population can be converted into tumor-specific effectors with clinical-grade antibodies, thus providing not only a therapeutic option for TNBC but also a valid alternative to more complex approaches based on chimeric antigen receptor-engineered cells. List of abbreviations ACT, Adoptive Cell Transfer; ADCC, Antibody-Dependent Cell-mediated Cytotoxicity; ADP, Adenosine diphosphate; BLI, Bioluminescence Imaging; CAR, Chimeric Antigen Receptor; CIK, Cytokine Induced Killer cells; CTX, Cetuximab; DMEM, Dulbecco’s Modified Eagle Medium; EGFR, Human Epidermal Growth Factor 1; ER, Estrogen; FBS, Fetal Bovine Serum; FFPE, Formalin-Fixed Paraffin-Embedded; GMP, Good Manufacturing Practices; GVHD, Graft Versus Host Disease; HER2, Human Epidermal Growth Factor 2; HRP, Horseradish Peroxidase; IFN-γ, Interferon-γ; IHC, Immunohistochemistry; IL-2, Interleukin-2; ISO, Irrelevant antibody; i.t., intratumoral; i.v., intravenous, mAbs, Monoclonal Antibodies; mIHC, Multiplex Fluorescence Immunohistochemistry; MHC, Major Histocompatibility Complex; NK, Natural Killer; NKG2D, Natural-Killer group 2 member D; NSG, NOD/SCID common γ chain knockout; PARP, Poly ADP-ribose polymerase; PBMCs, Peripheral Blood Mononuclear Cells; PBS, Phosphate-buffered saline; PDX, Patient-derived xenograft; PR, Progesterone; rhIFN-γ, Recombinant Human Interferon-γ; RPMI, Roswell Park Memorial Institute; STR, Short tandem Repeat; TCR, T Cell Receptor; TNBC, Triple Negative Breast Cancer; TSA, Tyramide Signal Amplification
Collapse
Affiliation(s)
| | - Elisa Cappuzzello
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy
| | - Anna Dalla Pietà
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy
| | - Anna Tosi
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy
| | - Pierangela Palmerini
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy
| | | | - Lorenzo Nicolè
- Department of Medicine, Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Antonio Rosato
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy.,Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy
| |
Collapse
|
10
|
Cancer Stem Cells in Soft-Tissue Sarcomas. Cells 2020; 9:cells9061449. [PMID: 32532153 PMCID: PMC7349510 DOI: 10.3390/cells9061449] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
Soft tissue sarcomas (STS) are a rare group of mesenchymal solid tumors with heterogeneous genetic profiles and clinical features. Systemic chemotherapy is the backbone treatment for advanced STS; however, STS frequently acquire resistance to standard therapies, which highlights the need to improve treatments and identify novel therapeutic targets. Increases in the knowledge of the molecular pathways that drive sarcomas have brought to light different molecular alterations that cause tumor initiation and progression. These findings have triggered a breakthrough of targeted therapies that are being assessed in clinical trials. Cancer stem cells (CSCs) exhibit mesenchymal stem cell (MSC) features and represent a subpopulation of tumor cells that play an important role in tumor progression, chemotherapy resistance, recurrence and metastasis. In fact, CSCs phenotypes have been identified in sarcomas, allied to drug resistance and tumorigenesis. Herein, we will review the published evidence of CSCs in STS, discussing the molecular characteristic of CSCs, the commonly used isolation techniques and the new possibilities of targeting CSCs as a way to improve STS treatment and consequently patient outcome.
Collapse
|
11
|
Capellero S, Erriquez J, Melano C, Mesiano G, Genta S, Pisacane A, Mittica G, Ghisoni E, Olivero M, Di Renzo MF, Aglietta M, Sangiolo D, Valabrega G. Preclinical immunotherapy with Cytokine-Induced Killer lymphocytes against epithelial ovarian cancer. Sci Rep 2020; 10:6478. [PMID: 32296104 PMCID: PMC7160190 DOI: 10.1038/s41598-020-63634-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 04/02/2020] [Indexed: 02/07/2023] Open
Abstract
Despite improvements in surgery and medical treatments, epithelial ovarian cancer (EOC) remains the most lethal gynaecological malignancy. Aim of this study is to investigate the preclinical immunotherapy activity of cytokine-induced killer lymphocytes (CIK) against epithelial ovarian cancers, focusing on platinum-resistant settings. We generated CIK ex vivo starting from human peripheral blood samples (PBMCs) collected from EOC patients. Their antitumor activity was tested in vitro and in vivo against platinum-resistant patient-derived ovarian cancer cells (pdOVCs) and a Patient Derived Xenograft (PDX), respectively. CIK were efficiently generated (48 fold median ex vivo expansion) from EOC patients; pdOVCs lines (n = 9) were successfully generated from metastatic ascites; the expression of CIK target molecules by pdOVC confirmed pre and post treatment in vitro with carboplatin. The results indicate that patient-derived CIK effectively killed autologous pdOVCs in vitro. Such intense activity was maintained against a subset of pdOVC that survived in vitro treatment with carboplatin. Moreover, CIK antitumor activity and tumor homing was confirmed in vivo within an EOC PDX model. Our preliminary data suggest that CIK are active in platinum resistant ovarian cancer models and should be therefore further investigated as a new therapeutic option in this extremely challenging setting.
Collapse
Affiliation(s)
- S Capellero
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - J Erriquez
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - C Melano
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - G Mesiano
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - S Genta
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - A Pisacane
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - G Mittica
- Unit of Oncology, ASL Verbano Cusio Ossola (VCO), Verbania, Italy
| | - E Ghisoni
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - M Olivero
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - M F Di Renzo
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - M Aglietta
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - D Sangiolo
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - G Valabrega
- Department of Oncology, University of Torino, Torino, Italy. .,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy.
| |
Collapse
|
12
|
Bremm M, Pfeffermann LM, Cappel C, Katzki V, Erben S, Betz S, Quaiser A, Merker M, Bonig H, Schmidt M, Klingebiel T, Bader P, Huenecke S, Rettinger E. Improving Clinical Manufacturing of IL-15 Activated Cytokine-Induced Killer (CIK) Cells. Front Immunol 2019; 10:1218. [PMID: 31214182 PMCID: PMC6554420 DOI: 10.3389/fimmu.2019.01218] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/13/2019] [Indexed: 12/18/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are an immunotherapeutic approach to combat relapse following allogeneic hematopoietic stem cell transplantation (HSCT) in acute leukemia or myelodysplastic syndrome (MDS) patients. Prompt and sequential administration of escalating cell doses improves the efficacy of CIK cell therapy without exacerbating graft vs. host disease (GVHD). This study addresses manufacturing-related issues and aimed to develop a time-, personal- and cost-saving good manufacturing process (GMP)-compliant protocol for the generation of ready-for-use therapeutic CIK cell doses starting from one unstimulated donor-derived peripheral blood (PB) or leukocytapheresis (LP) products. Culture medium with or without the addition of either AB serum, fresh frozen plasma (FFP) or platelet lysate (PL) was used for culture. Fresh and cryopreserved CIK cells were compared regarding expansion rate, viability, phenotype, and ability to inhibit leukemia growth. Cell numbers increased by a median factor of 10-fold in the presence of FFP, PL, or AB serum, whereas cultivation in FFP/PL-free or AB serum-free medium failed to promote adequate CIK cell proliferation (p < 0.01) needed to provide clinical doses of 1 × 106 T cells/kG, 5 × 106 T cells/kG, 1 × 107 T cells/kG, and 1 × 108 T cells/kG recipient body weight. CIK cells consisting of T cells, T- natural killer (T-NK) cells and a minor fraction of NK cells were not significantly modified by different medium supplements. Moreover, neither cytotoxic potential against leukemic THP-1 cells nor cell activation shown by CD25 expression were significantly influenced. Moreover, overnight and long-term cryopreservation had no significant effect on the composition of CIK cells, their phenotype or cytotoxic potential. A viability of almost 93% (range: 89–96) and 89.3% (range: 84–94) was obtained after freeze-thawing procedure and long-term storage, respectively, whereas viability was 96% (range: 90-97) in fresh CIK cells. Altogether, GMP-complaint CIK cell generation from an unstimulated donor-derived PB or LP products was feasible. Introducing FFP, which is easily accessible, into CIK cell cultures was time- and cost-saving without loss of viability and potency in a 10-12 day batch culture. The feasibility of cryopreservation enabled storage and delivery of sequential highly effective ready-for-use CIK cell doses and therefore reduced the number of manufacturing cycles.
Collapse
Affiliation(s)
- Melanie Bremm
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | | | - Claudia Cappel
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Verena Katzki
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Stephanie Erben
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Sibille Betz
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Andrea Quaiser
- Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Michael Merker
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Halvard Bonig
- Division for Translational Development of Cellular Therapeutics, Institute for Transfusion Medicine and Immunohematology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Michael Schmidt
- Division for Translational Development of Cellular Therapeutics, Institute for Transfusion Medicine and Immunohematology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Thomas Klingebiel
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Peter Bader
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Sabine Huenecke
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| | - Eva Rettinger
- Clinic for Pediatric and Adolescent Medicine, University Hospital, Frankfurt, Germany
| |
Collapse
|
13
|
Zhou Y, Chen CL, Jiang SW, Feng Y, Yuan L, Chen P, Zhang L, Huang S, Li J, Xia JC, Zheng M. Retrospective analysis of the efficacy of adjuvant CIK cell therapy in epithelial ovarian cancer patients who received postoperative chemotherapy. Oncoimmunology 2018; 8:e1528411. [PMID: 30713783 PMCID: PMC6343777 DOI: 10.1080/2162402x.2018.1528411] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/26/2018] [Accepted: 09/20/2018] [Indexed: 12/24/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are demonstrated to possess potent cytolytic effect against ovarian cancer cells in vitro and in vivo. However, the clinical efficacy of maintenance therapy of CIK cells in patients with epithelial ovarian cancer (EOC) after first-line treatment remains unclear. This retrospective study included 646 cases of postoperative EOC patients, 72 of which received chemotherapy and sequential immunotherapy (CIT group), and 574 of which received only chemotherapy (Control group). Patients in the CIT group received at least four cycles of CIK cell (range 8.0 × 109 – 1.3 × 1010 cells) transfusion, with the interval of each cycle being 2 weeks. Survival analysis showed a significantly higher overall survival (OS) rate in the CIT group compared with the control group, as well as a favorable progression-free survival (PFS). Univariate and multivariate analyses indicated that adjuvant CIT was an independent prognostic factor for the OS of patients with EOC. Furthermore, subgroup analyses showed that adjuvant CIT significantly improved the OS of patients older than 45 years, with CA125 ≤ 1000, or with moderate or poorly differentiated tumors, and prolonged the PFS of patients with residual disease > 1 cm. Additionally, Kaplan-Meier analyses revealed that a higher fraction of CD3+CD8+/CD3+CD56+ phenotypes or lower percentage of CD3+CD4+/CD3−CD56+ phenotypes in the infused CIK cells significantly associated with better survival of patients with EOC. Furthermore, across all processes of CIK cell immunotherapy in the CIT group, 12.5% (9/72) of patients developed self-limiting light fevers and shivering at grade 1 or 2. No immunotherapy-related serious reactions were recorded. These data indicate that adjuvant CIT with CIK cells is an effective therapeutic approach to prolonging the survival of EOC patients.
Collapse
Affiliation(s)
- Yun Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Gynecology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Chang-Long Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Radiation Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Sen-Wei Jiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Gynecology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yanling Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Gynecology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Linjing Yuan
- Department of Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Ping Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of VIP region, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Lan Zhang
- Department of Radiotherapy, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, Kunming, P. R. China
| | - Shuting Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Gynecology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jundong Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Gynecology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jian-Chuan Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Min Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Gynecology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| |
Collapse
|
14
|
Horenstein AL, Chillemi A, Zini R, Quarona V, Bianchi N, Manfredini R, Gambari R, Malavasi F, Ferrari D. Cytokine-Induced Killer Cells Express CD39, CD38, CD203a, CD73 Ectoenzymes and P1 Adenosinergic Receptors. Front Pharmacol 2018; 9:196. [PMID: 29731713 PMCID: PMC5920153 DOI: 10.3389/fphar.2018.00196] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/21/2018] [Indexed: 11/16/2022] Open
Abstract
Cytokine-induced killer (CIK) cells, a heterogeneous T cell population obtained by in vitro differentiation of peripheral blood mononuclear cells (PBMC), represent a promising immunological approach in cancer. Numerous studies have explored the role of CD38, CD39, CD203a/PC-1, and CD73 in generating extracellular adenosine (ADO) and thus in shaping the tumor niche in favor of proliferation. The findings shown here reveal that CIK cells are able to produce extracellular ADO via traditional (CD39/CD73) and/or alternative (CD38/CD203a/CD73 or CD203a/CD73) pathways. Transcriptome analysis showed the mRNA expression of these molecules and their modulation during PBMC to CIK differentiation. When PBMC from normal subjects or cancer bearing patients were differentiated into CIK cells under normoxic conditions, CD38 and CD39 were greatly up-regulated while the number of CD203a, and CD73 positive cells underwent minor changes. Since hypoxic conditions are often found in tumors, we asked whether CD39, CD38, CD203a, and CD73 expressed by CIK cells were modulated by hypoxia. PBMC isolated from cancer patients and differentiated into CIK cells in hypoxic conditions did not show relevant changes in CD38, CD39, CD73, CD203a, and CD26. CIK cells also expressed A1, A2A, and A2B ADO receptors and they only underwent minor changes as a consequence of hypoxia. The present study sheds light on a previously unknown functional aspect of CIK cells, opening the possibility of pharmacologically modulated ADO-generating ectoezymes to improve CIK cells performance.
Collapse
Affiliation(s)
- Alberto L Horenstein
- Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Antonella Chillemi
- Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Roberta Zini
- Centre for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Valeria Quarona
- Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Nicoletta Bianchi
- Department of Life Science and Biotechnology, Section of Microbiology and Applied Pathology, University of Ferrara, Ferrara, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto Gambari
- Department of Life Science and Biotechnology, Section of Microbiology and Applied Pathology, University of Ferrara, Ferrara, Italy
| | - Fabio Malavasi
- Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Davide Ferrari
- Department of Life Science and Biotechnology, Section of Microbiology and Applied Pathology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
15
|
Leuci V, Casucci GM, Grignani G, Rotolo R, Rossotti U, Vigna E, Gammaitoni L, Mesiano G, Fiorino E, Donini C, Pisacane A, Ambrosio LD, Pignochino Y, Aglietta M, Bondanza A, Sangiolo D. CD44v6 as innovative sarcoma target for CAR-redirected CIK cells. Oncoimmunology 2018; 7:e1423167. [PMID: 29721373 PMCID: PMC5927525 DOI: 10.1080/2162402x.2017.1423167] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/22/2017] [Accepted: 12/22/2017] [Indexed: 12/22/2022] Open
Abstract
Purpose of our study was to explore a new immunotherapy for high grade soft tissue sarcomas (STS) based on cytokine-induced killer cells (CIK) redirected with a chimeric antigen receptor (CAR) against the tumor-promoting antigen CD44v6. We aimed at generating bipotential killers, combining the CAR specificity with the intrinsic tumor-killing ability of CIK cells (CAR+.CIK). We set a patient-derived experimental platform. CAR+.CIK were generated by transduction of CIK precursors with a lentiviral vector encoding for anti-CD44v6-CAR. CAR+.CIK were characterized and assessed in vitro against multiple histotypes of patient-derived STS. The anti-sarcoma activity of CAR+.CIK was confirmed in a STS xenograft model. CD44v6 was expressed by 40% (11/27) of patient-derived STS. CAR+.CIK were efficiently expanded from patients (n = 12) and killed multiple histotypes of STS (including autologous targets, n = 4). The killing activity was significantly higher compared with unmodified CIK, especially at low effector/target (E/T) ratios: 98% vs 82% (E/T = 10:1) and 68% vs 26% (1:4), (p<0.0001). Specificity of tumor killing was confirmed by blocking with anti-CD44v6 antibody. CAR+.CIK produced higher amounts of IL6 and IFN-γ compared to control CIK. CAR+.CIK were highly active in mice bearing subcutaneous STS xenografts, with significant delay of tumor growth (p<0.0001) without toxicities. We report first evidence of CAR+.CIK's activity against high grade STS and propose CD44v6 as an innovative target in this setting. CIK are a valuable platform for the translation of CAR-based strategies to challenging field of solid tumors. Our findings support the exploration of CAR+.CIK in clinical trials against high grade STS.
Collapse
Affiliation(s)
- V Leuci
- Department of Oncology, University of Torino, Torino, Italy.,Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - G M Casucci
- Innovative Immunotherapies Unit, IRCCS San Raffaele Hospital Scientific Institute, Milano, Italy
| | - G Grignani
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - R Rotolo
- Department of Oncology, University of Torino, Torino, Italy
| | - U Rossotti
- Department of Oncology, University of Torino, Torino, Italy
| | - E Vigna
- Department of Oncology, University of Torino, Torino, Italy.,Laboratory of Gene Transfer, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - L Gammaitoni
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - G Mesiano
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - E Fiorino
- Department of Oncology, University of Torino, Torino, Italy
| | - C Donini
- Department of Oncology, University of Torino, Torino, Italy
| | - A Pisacane
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, (TO), Italy
| | - L D Ambrosio
- Department of Oncology, University of Torino, Torino, Italy.,Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - Y Pignochino
- Department of Oncology, University of Torino, Torino, Italy.,Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - M Aglietta
- Department of Oncology, University of Torino, Torino, Italy.,Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - A Bondanza
- Innovative Immunotherapies Unit, IRCCS San Raffaele Hospital Scientific Institute, Milano, Italy.,Vita-Salute San Raffaele University, Milano, Italy
| | - D Sangiolo
- Department of Oncology, University of Torino, Torino, Italy.,Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| |
Collapse
|
16
|
Abstract
INTRODUCTION Pancreatic cancer remains a deadly disease despite advances in surgery, chemotherapy, and radiation therapy. Treatment failure is likely due to intense chemoresistance and immunosuppression. Therefore, new treatment paradigms are urgently needed. Immunotherapy, particularly adoptive T cell transfer, is a highly-personalized therapy that involves the isolation and ex vivo expansion of tumor-specific T cells before administration to cancer-bearing hosts. Areas covered: This review summarizes different strategies of adoptive T cell therapy and their application in pancreatic cancer treatment. It also highlights recent advances and gives discussion on the future directions in T cell-based immunotherapy for pancreatic cancer. Expert opinion: Pancreatic ductal adenocarcinoma is extremely challenging to treat, in part, due to intense desmoplastic reaction and immunosuppression. The recent progress in cancer immunotherapy triggers a hope to use immunotherapeutic modality to treat pancreatic cancer. Immunotherapy is generally well tolerated, and has the potential to function as a monotherapy or in synergistic combination with conventional chemotherapy. We must make efforts to optimize the immunotherapeutic regimen and to select patients to treat based on their biological profile. To accomplish this goal, an intense collaboration is needed to bridge between bench and bedside.
Collapse
Affiliation(s)
- Fang Liu
- a GI Oncology Program and Experimental Therapeutics , Tufts University School of Medicine , Boston , MA , USA.,b PGY-2, Internal Medicine Residency Program at Metrowest Medical Center , Framingham , MA , USA
| | - Muhammad Wasif Saif
- a GI Oncology Program and Experimental Therapeutics , Tufts University School of Medicine , Boston , MA , USA
| |
Collapse
|
17
|
Generation and characterization of ErbB2-CAR-engineered cytokine-induced killer cells for the treatment of high-risk soft tissue sarcoma in children. Oncotarget 2017; 8:66137-66153. [PMID: 29029499 PMCID: PMC5630399 DOI: 10.18632/oncotarget.19821] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 06/29/2017] [Indexed: 01/03/2023] Open
Abstract
Pediatric patients with recurrent, refractory or advanced soft tissue sarcoma (STS) who are simultaneously showing signs of cumulative treatment toxicity are in need of novel therapies. In this preclinical analysis, we identified ErbB2 as a targetable antigen on STS cells and used cytokine-induced killer (CIK) cells transduced with the lentiviral 2nd-generation chimeric antigen receptor (CAR) vector pS-5.28.z-IEW to target ErbB2-positive tumors. Solely CIK cell subsets with the CD3+ T cell phenotype showed up to 85% cell surface expression of the respective CAR. A comparison of wildtype (WT), mock-vector and ErbB2-CAR-CIK cells showed, that engineered cells exhibited diminished in vitro expansion, retained WT CIK cell phenotype with higher percentages of differentiated effector memory/effector cells. Activating natural killer (NK) cell receptor NKG2D-restricted target cell recognition and killing of WT and ErbB2-CAR-CIK cells was maintained against ErbB2-negative tumors, while ErbB2-CAR-CIK cells demonstrated significantly increased cytotoxicity against ErbB2-positive targets, including primary tumors. ErbB2-CAR- but not WT CIK cells proliferated, infiltrated and efficiently lysed tumor cell monolayers as well as 3D tumor spheroids. Here, we demonstrate a potential cell therapeutic approach using ErbB2-CAR-CIK cells for the recognition and elimination of tumor cells expressing ErbB2, which we identified as a targetable antigen on high-risk STS cells.
Collapse
|
18
|
Han D, Rodriguez-Bravo V, Charytonowicz E, Demicco E, Domingo-Domenech J, Maki RG, Cordon-Cardo C. Targeting sarcoma tumor-initiating cells through differentiation therapy. Stem Cell Res 2017; 21:117-123. [PMID: 28433655 DOI: 10.1016/j.scr.2017.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/23/2017] [Accepted: 04/11/2017] [Indexed: 01/31/2023] Open
Abstract
Human leukocyte antigen class I (HLA-I) down-regulation has been reported in many human cancers to be associated with poor clinical outcome. However, its connection to tumor-initiating cells (TICs) remains unknown. In this study, we report that HLA-I is down-regulated in a subpopulation of cells that have high tumor initiating capacity in different types of human sarcomas. Detailed characterization revealed their distinct molecular profiles regarding proliferation, apoptosis and stemness programs. Notably, these TICs can be induced to differentiate along distinct mesenchymal lineages, including the osteogenic pathway. The retinoic acid receptor signaling pathway is overexpressed in HLA-1 negative TICs. All-trans retinoic acid treatment successfully induced osteogenic differentiation of this subpopulation, in vitro and in vivo, resulting in significantly decreased tumor formation. Thus, our findings indicate down-regulated HLA-I is a shared feature of TICs in a variety of human sarcomas, and differentiation therapy strategies may specifically target undifferentiated TICs and inhibit tumor formation.
Collapse
Affiliation(s)
- Dan Han
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | | | - Elizabeth Demicco
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Josep Domingo-Domenech
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert G Maki
- Department of Hematology-Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carlos Cordon-Cardo
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
19
|
Liu D, Staveley-O’Carroll KF, Li G. Immune-based Therapy Clinical Trials in Hepatocellular Carcinoma. JOURNAL OF CLINICAL & CELLULAR IMMUNOLOGY 2015; 6:376. [PMID: 26877890 PMCID: PMC4750497 DOI: 10.4172/2155-9899.1000376] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related mortality and continues to increase. Current standard of care for patients with HCC only provides limited therapeutic benefit. Development of innovative strategies is urgently needed. Experience with immunotherapy in HCC is quite early, but rapidly rise in the recent 15 years. Multifaceted immune-based approaches have shown efficacy in achieving disease regression, representing the most promising new treatment approach. Here, we classify the ongoing or completed clinical trials in HCC in terms of the immune strategies to be used and assess their clinical outcomes. The generated information may be helpful in the design of future immune-based therapies for achieving ideal tumor control and maximizing anti-tumor immunity.
Collapse
Affiliation(s)
- Dai Liu
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kevin F. Staveley-O’Carroll
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| | - Guangfu Li
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|