1
|
Hato L, Vizcay A, Eguren I, Pérez-Gracia JL, Rodríguez J, Gállego Pérez-Larraya J, Sarobe P, Inogés S, Díaz de Cerio AL, Santisteban M. Dendritic Cells in Cancer Immunology and Immunotherapy. Cancers (Basel) 2024; 16:981. [PMID: 38473341 DOI: 10.3390/cancers16050981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/15/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Cancer immunotherapy modulates the immune system, overcomes immune escape and stimulates immune defenses against tumors. Dendritic cells (DCs) are professional promoters of immune responses against tumor antigens with the outstanding ability to coordinate the innate and adaptive immune systems. Evidence suggests that there is a decrease in both the number and function of DCs in cancer patients. Therefore, they represent a strong scaffold for therapeutic interventions. DC vaccination (DCV) is safe, and the antitumoral responses induced are well established in solid tumors. Although the addition of checkpoint inhibitors (CPIs) to chemotherapy has provided new options in the treatment of cancer, they have shown no clinical benefit in immune desert tumors or in those tumors with dysfunctional or exhausted T-cells. In this way, DC-based therapy has demonstrated the ability to modify the tumor microenvironment for immune enriched tumors and to potentiate systemic host immune responses as an active approach to treating cancer patients. Application of DCV in cancer seeks to obtain long-term antitumor responses through an improved T-cell priming by enhancing previous or generating de novo immune responses. To date, DCV has induced immune responses in the peripheral blood of patients without a significant clinical impact on outcome. Thus, improvements in vaccines formulations, selection of patients based on biomarkers and combinations with other antitumoral therapies are needed to enhance patient survival. In this work, we review the role of DCV in different solid tumors with their strengths and weaknesses, and we finally mention new trends to improve the efficacy of this immune strategy.
Collapse
Affiliation(s)
- Laura Hato
- Immunology, Riberalab, 03203 Alicante, Spain
| | - Angel Vizcay
- Medical Oncology, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | - Iñaki Eguren
- Medical Oncology, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | | | - Javier Rodríguez
- Medical Oncology, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | | | - Pablo Sarobe
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
- Program of Immunology and Immunotherapy, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, 31008 Pamplona, Spain
- CIBEREHD, 31008 Pamplona, Spain
| | - Susana Inogés
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
- Cell Therapy Unit, Program of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Ascensión López Díaz de Cerio
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
- Cell Therapy Unit, Program of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Marta Santisteban
- Medical Oncology, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| |
Collapse
|
2
|
Pandey S, Cholak ME, Yadali R, Sosman JA, Tetreault MP, Fang D, Pollack SM, Gnjatic S, Obeng RC, Lyerly HK, Sonabend AM, Guevara-Patiño JA, Butterfield LH, Zhang B, Maecker HT, Le Poole IC. Immune Assessment Today: Optimizing and Standardizing Efforts to Monitor Immune Responses in Cancer and Beyond. Cancers (Basel) 2024; 16:475. [PMID: 38339227 PMCID: PMC10854499 DOI: 10.3390/cancers16030475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 01/16/2024] [Indexed: 02/12/2024] Open
Abstract
As part of a symposium, current and former directors of Immune Monitoring cores and investigative oncologists presented insights into the past, present and future of immune assessment. Dr. Gnjatic presented a classification of immune monitoring technologies ranging from universally applicable to experimental protocols, while emphasizing the need for assay harmonization. Dr. Obeng discussed physiologic differences among CD8 T cells that align with anti-tumor responses. Dr. Lyerly presented the Soldano Ferrone lecture, commemorating the passionate tumor immunologist who inspired many, and covered a timeline of monitoring technology development and its importance to immuno-oncology. Dr. Sonabend presented recent achievements in glioblastoma treatment, accentuating the range of monitoring techniques that allowed him to refine patient selection for clinical trials. Dr. Guevara-Patiño focused on hypoxia within the tumor environment and stressed that T cell viability is not to be confused with functionality. Dr. Butterfield accentuated monitoring of dendritic cell metabolic (dys)function as a determinant for tumor vaccine success. Lectures were interspersed with select abstract presentations. To summarize the concepts, Dr. Maecker from Stanford led an informative forum discussion, pointing towards the future of immune monitoring. Immune monitoring continues to be a guiding light towards effective immunotherapeutic strategies.
Collapse
Affiliation(s)
- Surya Pandey
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
| | - Meghan E. Cholak
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
| | - Rishita Yadali
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
| | - Jeffrey A. Sosman
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Marie-Pier Tetreault
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Deyu Fang
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Seth M. Pollack
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Sacha Gnjatic
- Human Immune Monitoring Center, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Rebecca C. Obeng
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - H. Kim Lyerly
- Center for Applied Therapeutics, Duke Cancer Center, Duke University, Durham, NC 27710, USA;
| | - Adam M. Sonabend
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | | | - Lisa H. Butterfield
- Merck Research Laboratories, Boston, MA 02115, USA;
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143, USA
| | - Bin Zhang
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Holden T. Maecker
- Human Immune Monitoring Center, Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - I. Caroline Le Poole
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| |
Collapse
|
3
|
Berk E, Kalinski P. Lymphocyte-polarized DC1s: Effective inducers of tumor-specific CTLs. Oncoimmunology 2021; 1:1443-1444. [PMID: 23243623 PMCID: PMC3518532 DOI: 10.4161/onci.21295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Activated lymphocytes secrete dendritic cell (DC)-activating cytokines including tumor necrosis factor α and interferon γ, and induce Type-1-polarized DCs (DC1s). Lymphocyte-polarized DC1s secrete high levels of biologically active interleukin-12 (IL-12p70) and CXCL10 and show enhanced CTL-inducing activity. Our data demonstrate the feasibility of using autologous lymphocytes to enhance the immunogenic properties of DCs in a low-cost clinically-compatible process.
Collapse
Affiliation(s)
- Erik Berk
- Department of Sugery; University of Pittsburgh; Pittsburgh, PA USA
| | | |
Collapse
|
4
|
Ramanathan R, Choudry H, Jones H, Girgis M, Gooding W, Kalinski P, Bartlett DL. Phase II Trial of Adjuvant Dendritic Cell Vaccine in Combination with Celecoxib, Interferon-α, and Rintatolimod in Patients Undergoing Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy for Peritoneal Metastases. Ann Surg Oncol 2021; 28:4637-4646. [PMID: 33400000 PMCID: PMC7784622 DOI: 10.1245/s10434-020-09464-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/25/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Peritoneal metastases portend poor prognosis in the setting of standard chemotherapy. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS/HIPEC) improves outcomes, but relapse is common. We report a phase II trial evaluating the safety and efficacy of adjuvant αDC1 vaccination with chemokine modulation (CKM) after CRS/HIPEC. METHODS Patients undergoing CRS/HIPEC for appendiceal cancer, colorectal cancer, or peritoneal mesothelioma were enrolled. In addition to standard adjuvant chemotherapy, patients received intranodal and intradermal injections of autologous tumor-loaded αDC1 vaccine. After each vaccine booster, patients received CKM over 4 days, consisting of celecoxib, interferon (IFN)-α, and rintatolimod. RESULTS Forty-six patients underwent CRS/HIPEC followed by αDC1 treatment, including 24 appendiceal primaries, 20 colorectal, and 2 mesotheliomas. DC maturation was successful, with 97% expressing HLA-DR and CD86. Tumor cell recovery from peritoneal tumors was challenging, resulting in only 17% of patients receiving the target dose of αDC1. The αDC1 and CKM regimen was well tolerated. CKM successfully modulated serum inflammatory cytokine and chemokine levels. Median progression-free survival (PFS) for appendiceal primaries was 50.4, 34.2, and 8.9 months for grade 1, 2, and 3 tumors, respectively, while median PFS for colorectal cancer was 20.5 and 8.9 months for moderately and poorly differentiated tumors, respectively. CONCLUSIONS Adjuvant autologous tumor antigen-loaded αDC1 vaccine and CKM is well tolerated. The mucinous nature of peritoneal metastases limits the feasibility of obtaining adequate autologous tumor cells. The improvement in median PFS did not meet our predefined thresholds, leading us to conclude that αDC1 vaccination is not appropriate for patients undergoing CRS/HIPEC for peritoneal metastases.
Collapse
Affiliation(s)
- Rajesh Ramanathan
- Division of Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.,Department of Surgery, Banner MD Anderson Cancer Center, Phoenix, AZ, USA
| | - Haroon Choudry
- Division of Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Heather Jones
- Division of Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Mark Girgis
- Division of Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.,Department of Surgery, UCLA Health, Los Angeles, CA, USA
| | - William Gooding
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Pawel Kalinski
- Medical Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - David L Bartlett
- Division of Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA. .,Department of Surgery, AHN Cancer Institute, Pittsburgh, PA, USA.
| |
Collapse
|
5
|
Butterfield LH, Vujanovic L, Santos PM, Maurer DM, Gambotto A, Lohr J, Li C, Waldman J, Chandran U, Lin Y, Lin H, Tawbi HA, Tarhini AA, Kirkwood JM. Multiple antigen-engineered DC vaccines with or without IFNα to promote antitumor immunity in melanoma. J Immunother Cancer 2019; 7:113. [PMID: 31014399 PMCID: PMC6480917 DOI: 10.1186/s40425-019-0552-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 02/27/2019] [Indexed: 02/08/2023] Open
Abstract
Background Cancer vaccines are designed to promote systemic antitumor immunity and tumor eradication. Cancer vaccination may be more efficacious in combination with additional interventions that may build on or amplify their effects. Methods Based on our previous clinical and in vitro studies, we designed an antigen-engineered DC vaccine trial to promote a polyclonal CD8+ and CD4+ T cell response against three shared melanoma antigens. The 35 vaccine recipients were then randomized to receive one month of high-dose IFNα or observation. Results The resulting clinical outcomes were 2 partial responses, 8 stable disease and 14 progressive disease among patients with measurable disease using RECIST 1.1, and, of 11 surgically treated patients with no evidence of disease (NED), 4 remain NED at a median follow-up of 3 years. The majority of vaccinated patients showed an increase in vaccine antigen-specific CD8+ and CD4+ T cell responses. The addition of IFNα did not appear to improve immune or clinical responses in this trial. Examination of the DC vaccine profiles showed that IL-12p70 secretion did not correlate with immune or clinical responses. In depth immune biomarker studies support the importance of circulating Treg and MDSC for development of antigen-specific T cell responses, and of circulating CD8+ and CD4+ T cell subsets in clinical responses. Conclusions DC vaccines are a safe and reliable platform for promoting antitumor immunity. This combination with one month of high dose IFNα did not improve outcomes. Immune biomarker analysis in the blood identified several predictive and prognostic biomarkers for further analysis, including MDSC. Trial registration NCT01622933. Electronic supplementary material The online version of this article (10.1186/s40425-019-0552-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lisa H Butterfield
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA. .,Department of Surgery, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA. .,Department of Immunology, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA. .,UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.
| | - Lazar Vujanovic
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Patricia M Santos
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Deena M Maurer
- Department of Immunology, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Andrea Gambotto
- Department of Surgery, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Joel Lohr
- Department of Immunology, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Chunlei Li
- UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,Present address: Tsinghua University School of Medicine, Beijing, China
| | - Jacob Waldman
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Uma Chandran
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yan Lin
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Huang Lin
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hussein A Tawbi
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,Present address: Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ahmad A Tarhini
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,Present address: Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - John M Kirkwood
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| |
Collapse
|
6
|
Garcia-Bates TM, Palma ML, Shen C, Gambotto A, Macatangay BJC, Ferris RL, Rinaldo CR, Mailliard RB. Contrasting Roles of the PD-1 Signaling Pathway in Dendritic Cell-Mediated Induction and Regulation of HIV-1-Specific Effector T Cell Functions. J Virol 2019; 93:e02035-18. [PMID: 30541848 PMCID: PMC6384070 DOI: 10.1128/jvi.02035-18] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 11/30/2018] [Indexed: 12/25/2022] Open
Abstract
Eliciting highly functional CD8+ cytotoxic T lymphocyte (CTL) responses against a broad range of epitopes will likely be required for immunotherapeutic control of HIV-1 infection. However, the combination of CTL exhaustion and the ability of HIV-1 to rapidly establish CTL escape variants presents major hurdles toward this goal. Our previous work highlighted the use of monocyte-derived, mature, high-interleukin-12 (IL-12)-producing type 1 polarized dendritic cells (MDC1) to selectively induce more potent effector CTLs derived from naive, rather than memory, CD8+ T cell precursors isolated from HIV-1-positive participants in the Multicenter AIDS Cohort Study. In this study, we report that these highly stimulatory antigen-presenting cells also express enhanced levels of the coinhibitory molecule programmed cell death ligand 1 (PD-L1), the ligand for PD-1, which is further upregulated upon subsequent stimulation with the CD4+ T helper cell-derived factor CD40L. Interestingly, blocking the PD-1 signaling pathway during MDC1 induction of HIV-1-specific CTL responses inhibited the priming, activation, and differentiation of naive CD8+ T cells into effector T cells expressing high levels of T-box transcription factor (T-bethi) and eomesodermin (Eomes+). In contrast, PD-1 blockade enhanced the overall magnitude of memory HIV-specific CTL responses and reversed the exhausted memory phenotype from a T-betlow/Eomes+ to a T-bethi/Eomes+ phenotype. These results indicate that the PD-L1/PD-1 signaling pathway has a previously unappreciated dual role in the induction and regulation of HIV-1-specific CTL immunity, which is greatly determined by the context and differentiation stage of the responsive CD8+ T cells.IMPORTANCE Targeting the PD-1/PD-L1 immune checkpoint axis with signaling inhibitors has proven to be a powerful immunotherapeutic strategy to enhance the functional quality and survival of existing antigen-specific effector T cells. However, our study demonstrates that the context and timing of PD-1 signaling in T cells greatly impact the outcome of the effector response. In particular, we show that PD-1 activation plays a positive role during the DC-mediated initiation stage of the primary T cell response, while it serves as an inhibitory mechanism during the effector phase of the response. Therefore, caution should be taken in the design of therapies that include targeting of the PD-1/PD-L1 signaling pathway in order to avoid potential negative impacts on the induction of de novo T cell responses.
Collapse
Affiliation(s)
- Tatiana M Garcia-Bates
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Mariana L Palma
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Chengli Shen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Andrea Gambotto
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bernard J C Macatangay
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert L Ferris
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Charles R Rinaldo
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robbie B Mailliard
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
DE Wolf C, VAN DE Bovenkamp M, Hoefnagel M. Regulatory perspective on in vitro potency assays for human dendritic cells used in anti-tumor immunotherapy. Cytotherapy 2018; 20:1289-1308. [PMID: 30327247 DOI: 10.1016/j.jcyt.2018.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/25/2018] [Accepted: 07/14/2018] [Indexed: 12/18/2022]
Abstract
Dendritic cells (DCs) are key connectors between the innate and adaptive immune system and have an important role in modulating other immune cells. Therefore, their therapeutic application to steer immune responses is considered in various disorders, including cancer. Due to differences in the cell source and manufacturing process, each DC medicinal product is unique. Consequently, release tests to ensure consistent quality need to be product-specific. Although general guidance concerning quality control testing of cell-based therapies is available, cell type-specific regulation is still limited. Especially guidance related to potency testing is needed, because developing an in vitro assay measuring cell properties relevant for in vivo functionality is challenging. In this review, we provide DC-specific guidance for development of in vitro potency assays for characterisation and release. We present a broad overview of in vitro potency assays suggested for DC products to determine their anti-tumor functionality. Several advantages and limitations of these assays are discussed. Also, we provide some points to consider for selection and design of a potency test. The ideal functionality assay for anti-tumor products evaluates the capacity of DCs to stimulate antigen-specific T cells. Because this approach may not be feasible for release, use of surrogate potency markers could be considered, provided that these markers are sufficiently linked to the in vivo DC biological activity and clinical response. Further elucidation of the involvement of specific DC subsets in anti-tumor responses will result in improved manufacturing processes for DC-based products and should be considered during potency assay development.
Collapse
Affiliation(s)
- Charlotte DE Wolf
- Medicines Evaluation Board College ter Beoordeling van Geneesmiddelen-Medicines Evaluation Board (CBG-MEB), Utrecht, The Netherlands; Department of Infectious Diseases and Immunology, Utrecht University, The Netherlands
| | - Marja VAN DE Bovenkamp
- Medicines Evaluation Board College ter Beoordeling van Geneesmiddelen-Medicines Evaluation Board (CBG-MEB), Utrecht, The Netherlands
| | - Marcel Hoefnagel
- Medicines Evaluation Board College ter Beoordeling van Geneesmiddelen-Medicines Evaluation Board (CBG-MEB), Utrecht, The Netherlands.
| |
Collapse
|
8
|
Obermajer N, Urban J, Wieckowski E, Muthuswamy R, Ravindranathan R, Bartlett DL, Kalinski P. Promoting the accumulation of tumor-specific T cells in tumor tissues by dendritic cell vaccines and chemokine-modulating agents. Nat Protoc 2018; 13:335-357. [PMID: 29345636 DOI: 10.1038/nprot.2017.130] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This protocol describes how to induce large numbers of tumor-specific cytotoxic T cells (CTLs) in the spleens and lymph nodes of mice receiving dendritic cell (DC) vaccines and how to modulate tumor microenvironments (TMEs) to ensure effective homing of the vaccination-induced CTLs to tumor tissues. We also describe how to evaluate the numbers of tumor-specific CTLs within tumors. The protocol contains detailed information describing how to generate a specialized DC vaccine with augmented ability to induce tumor-specific CTLs. We also describe methods to modulate the production of chemokines in the TME and show how to quantify tumor-specific CTLs in the lymphoid organs and tumor tissues of mice receiving different treatments. The combined experimental procedure, including tumor implantation, DC vaccine generation, chemokine-modulating (CKM) approaches, and the analyses of tumor-specific systemic and intratumoral immunity is performed over 30-40 d. The presented ELISpot-based ex vivo CTL assay takes 6 h to set up and 5 h to develop. In contrast to other methods of evaluating tumor-specific immunity in tumor tissues, our approach allows detection of intratumoral T-cell responses to nonmanipulated weakly immunogenic cancers. This detection method can be performed using basic laboratory skills, and facilitates the development and preclinical evaluation of new immunotherapies.
Collapse
Affiliation(s)
- Nataša Obermajer
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Julie Urban
- Immunotransplantation Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Eva Wieckowski
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Immunotransplantation Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | - David L Bartlett
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Pawel Kalinski
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Immunotransplantation Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
9
|
Akasaki Y, Kikuchi T, Homma S, Koido S, Ohkusa T, Tasaki T, Hayashi K, Komita H, Watanabe N, Suzuki Y, Yamamoto Y, Mori R, Arai T, Tanaka T, Joki T, Yanagisawa T, Murayama Y. Phase I/II trial of combination of temozolomide chemotherapy and immunotherapy with fusions of dendritic and glioma cells in patients with glioblastoma. Cancer Immunol Immunother 2016; 65:1499-1509. [PMID: 27688162 PMCID: PMC11028634 DOI: 10.1007/s00262-016-1905-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 09/22/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND This trial was designed to evaluate the safety and clinical responses to a combination of temozolomide (TMZ) chemotherapy and immunotherapy with fusions of DCs and glioma cells in patients with glioblastoma (GBM). METHOD GBM patients were assigned to two groups: a group of recurrent GBMs after failing TMZ-chemotherapy against the initially diagnosed glioma (Group-R) or a group of newly diagnosed GBMs (Group-N). Autologous cultured glioma cells obtained from surgical specimens were fused with autologous DCs using polyethylene glycol. The fusion cells (FC) were inoculated intradermally in the cervical region. Toxicity, progression-free survival (PFS), and overall survival (OS) of this trial were evaluated. Expressions of WT-1, gp-100, and MAGE-A3, recognized as chemoresistance-associated peptides (CAP), were confirmed by immunohistochemistry of paraffin-embedded tumor samples. Patient's PBMCs of pre- and post-vaccination were evaluated by tetramer and ELISPOT assays. RESULTS FC-immunotherapy was well tolerated in all patients. Medians of PFS and OS of Group-R (n = 10) were 10.3 and 18.0 months, and those of Group-N (n = 22) were 18.3 and 30.5 months, respectively. Up-regulation and/or cytoplasmic accumulation of CAPs was observed in the recurrent tumors of Group-R patients compared with their initially excised tumors. Specific immune responses against CAPs were observed in the tetramer and ELISPOT assays. CONCLUSIONS The combination of TMZ-treatment leading to up-regulation and/or cytoplasmic accumulation of CAPs, with FC-immunotherapy as a means of producing specific immunity against CAPs, may safely induce anti-tumor effects in patients with GBM.
Collapse
Affiliation(s)
- Yasuharu Akasaki
- Department of Neurosurgery, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan.
| | - Tetsuro Kikuchi
- Division of Oncology, Research Center for Medical Science, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Sadamu Homma
- Division of Oncology, Research Center for Medical Science, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Shigeo Koido
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Toshifumi Ohkusa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Tetsunori Tasaki
- Division of Blood Transfusion, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Kazumi Hayashi
- Division of Oncology, Research Center for Medical Science, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Hideo Komita
- Division of Oncology, Research Center for Medical Science, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Nobuyuki Watanabe
- Department of Neurosurgery, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yuta Suzuki
- Department of Neurosurgery, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yohei Yamamoto
- Department of Neurosurgery, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Ryosuke Mori
- Department of Neurosurgery, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Takao Arai
- Department of Neurosurgery, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Toshihide Tanaka
- Department of Neurosurgery, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Tatsuhiro Joki
- Department of Neurosurgery, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Takaaki Yanagisawa
- Department of Neurosurgery, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yuichi Murayama
- Department of Neurosurgery, Jikei University School of Medicine, 3-25-8 Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| |
Collapse
|
10
|
Pulsed Dendritic Cells for the Therapy of Experimental Glioma. Bull Exp Biol Med 2016; 161:792-796. [DOI: 10.1007/s10517-016-3512-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Indexed: 12/12/2022]
|
11
|
Bernal-Estévez D, Sánchez R, Tejada RE, Parra-López C. Chemotherapy and radiation therapy elicits tumor specific T cell responses in a breast cancer patient. BMC Cancer 2016; 16:591. [PMID: 27484900 PMCID: PMC4971722 DOI: 10.1186/s12885-016-2625-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/26/2016] [Indexed: 12/14/2022] Open
Abstract
Background Experimental evidence and clinical studies in breast cancer suggest that some anti-tumor therapy regimens generate stimulation of the immune system that accounts for tumor clinical responses, however, demonstration of the immunostimulatory power of these therapies on cancer patients continues to be a formidable challenge. Here we present experimental evidence from a breast cancer patient with complete clinical response after 7 years, associated with responsiveness of tumor specific T cells. Methods T cells were obtained before and after anti-tumor therapy from peripheral blood of a 63-years old woman diagnosed with ductal breast cancer (HER2/neu+++, ER-, PR-, HLA-A*02:01) treated with surgery, followed by paclitaxel, trastuzumab (suspended due to cardiac toxicity), and radiotherapy. We obtained a leukapheresis before surgery and after 8 months of treatment. Using in vitro cell cultures stimulated with autologous monocyte-derived dendritic cells (DCs) that produce high levels of IL-12, we characterize by flow cytometry the phenotype of tumor associated antigens (TAAs) HER2/neu and NY-ESO 1 specific T cells. The ex vivo analysis of the TCR-Vβ repertoire of TAA specific T cells in blood and Tumor Infiltrating Lymphocytes (TILs) were performed in order to correlate both repertoires prior and after therapy. Results We evidence a functional recovery of T cell responsiveness to polyclonal stimuli and expansion of TAAs specific CD8+ T cells using peptide pulsed DCs, with an increase of CTLA-4 and memory effector phenotype after anti-tumor therapy. The ex vivo analysis of the TCR-Vβ repertoire of TAA specific T cells in blood and TILs showed that whereas the TCR-Vβ04-02 clonotype is highly expressed in TILs the HER2/neu specific T cells are expressed mainly in blood after therapy, suggesting that this particular TCR was selectively enriched in blood after anti-tumor therapy. Conclusions Our results show the benefits of anti-tumor therapy in a breast cancer patient with clinical complete response in two ways, by restoring the responsiveness of T cells by increasing the frequency and activation in peripheral blood of tumor specific T cells present in the tumor before therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2625-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David Bernal-Estévez
- Immunology and Traslational Medicine Research Group, Graduated School in Biomedical Sciences, Department of Microbiology, Medical School, Universidad Nacional de Colombia, Carrera 30 #45-03 Building 471, office 304, Bogotá, Colombia South-America.,Immunology and Clinical Oncology Research Group (GIIOC), Fundación Salud de los Andes, Calle 44 No. 58-05, Bogotá, Colombia South-America
| | - Ramiro Sánchez
- Clínica del Seno, Carrera 11 # 68-36, Bogotá, Colombia South-America
| | - Rafael E Tejada
- Hospital Occidente de Kennedy E.S.E., Servicio de Oncología, Bogotá, Colombia South-America
| | - Carlos Parra-López
- Immunology and Traslational Medicine Research Group, Graduated School in Biomedical Sciences, Department of Microbiology, Medical School, Universidad Nacional de Colombia, Carrera 30 #45-03 Building 471, office 304, Bogotá, Colombia South-America. .,Facultad de Medicina, Departamento de Microbiología, Universidad Nacional de Colombia, Carrera 30 Calle 45, Bogotá, Colombia.
| |
Collapse
|
12
|
van den Bos C, Keefe R, Schirmaier C, McCaman M. Therapeutic human cells: manufacture for cell therapy/regenerative medicine. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2016; 138:61-97. [PMID: 23934363 DOI: 10.1007/10_2013_233] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
: Human primary cells (e.g. adult stem cells) as well as differentiated cells, including those of the immune system, have been found to be therapeutically useful and free of ethical concerns. Several products have received market authorization and numerous promising clinical trials are underway. We believe that such primary therapeutic cells will dominate the market for cell therapy applications for the foreseeable future. Consequently, production of such cellular products warrants attention and needs to be a fully controlled pharmaceutical process. Thus, where possible, such production should change from manufacture towards a truly scalable industrialized process for both allogeneic and autologous products. Here, we discuss manufacturing aspects of both autogeneic and allogeneic products, review the field, and provide historical context.
Collapse
|
13
|
Radomski M, Zeh HJ, Edington HD, Pingpank JF, Butterfield LH, Whiteside TL, Wieckowski E, Bartlett DL, Kalinski P. Prolonged intralymphatic delivery of dendritic cells through implantable lymphatic ports in patients with advanced cancer. J Immunother Cancer 2016; 4:24. [PMID: 27096100 PMCID: PMC4835859 DOI: 10.1186/s40425-016-0128-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 04/01/2016] [Indexed: 12/03/2022] Open
Abstract
Background The currently-used modes of administration of immunotherapeutic agents result in their limited delivery to the lymph nodes and/or require repetitive ultrasound-guided nodal injections or microsurgical lymphatic injections, limiting their feasibility. Here, we report on the feasibility and safety of a new method of long-term repetitive intralymphatic (IL) infusion of immune cells, using implantable delivery ports. Methods Nine patients with stage IV recurrent colorectal cancer underwent complete resection and received autologous dendritic cells (DCs) loaded with killed autologous tumor cells, KLH and PADRE, for up to four monthly cycles. Leg lymphatic vessels were cannulated, connected to 6.6Fr low-profile implantable subcutaneous delivery ports, and used to infuse 12 doses of DC over each 72 h-long cycle (every 6 h), followed by heparin flushes of the cannula-port system (one 72 h-long cycle per month). The patients who opted for alternative route of vaccine administration (2 patients) or whose ports became non-functional between cycles, continued treatment via intranodal (one injection/cycle) or intradermal (four injections/cycle) routes. Results A total of nine lymphatic cannulations and implantations of subcutaneous delivery ports were attempted in seven patients, with a success rate of eight out of nine (89 %). The average patency of the IL delivery system was 7.5 (±3.2) weeks. All six patients with IL ports successfully completed at least one complete 72 h-long DC infusion cycle (12 injections). Five patients (56 %) completed two full IL cycles (24 IL injections). No patients received more than two IL cycles without replacement of the IL port, due to catheter occlusion and/or local side effects: cellulitis and hematoma. Intranodal and intradermal backup options were used in, respectively, one and two patients. Overall cohort survival was >28 (±25) months. One patient with aggressive recurrent carcinomatosis, who received DC vaccines by intranodal route is alive at > 90 months, without evidence of disease. Conclusions We conclude that an intermediate-duration IL delivery of multiple doses of immunotherapeutic factors using implantable delivery ports is feasible, highly-tolerable and can be reproducibly performed in cancer patients to administer immune cells, or potentially, other immune factors. However, long-term IL port placement (>7.5 weeks), is not a currently-feasible option. Trial registration NCT00558051, registered Nov. 13, 2007.
Collapse
Affiliation(s)
- Michal Radomski
- Department of Surgery, Division of Surgical Oncology, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 400, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA
| | - Herbert J Zeh
- Department of Surgery, Division of Surgical Oncology, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 400, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA ; University of Pittsburgh Cancer Institute, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 500, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA
| | - Howard D Edington
- Department of Surgery, Division of Surgical Oncology, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 400, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA ; University of Pittsburgh Cancer Institute, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 500, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA
| | - James F Pingpank
- Department of Surgery, Division of Surgical Oncology, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 400, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA ; University of Pittsburgh Cancer Institute, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 500, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA
| | - Lisa H Butterfield
- Department of Surgery, Division of Surgical Oncology, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 400, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA ; University of Pittsburgh Cancer Institute, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 500, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA ; Department of Medicine, Hillman Cancer Center, UPCI Research Pavilion, Suite 137, 5117 Centre Avenue, Pittsburgh, PA 15213-1863 USA ; Department of Immunology, Hillman Cancer Center, UPCI Research Pavilion, Suite 1.46, 5117 Centre Avenue, Pittsburgh, PA 15213-1863 USA
| | - Theresa L Whiteside
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 500, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA ; Department of Immunology, Hillman Cancer Center, UPCI Research Pavilion, Suite 1.46, 5117 Centre Avenue, Pittsburgh, PA 15213-1863 USA ; Department of Pathology, Hillman Cancer Center, UPCI Research Pavilion, Suite 132, 5117 Centre Avenue, Pittsburgh, PA 15213-1863 USA ; Department of Otolaryngology, Hillman Cancer Center, UPCI Research Pavilion, Suite 132, 5117 Centre Avenue, Pittsburgh, PA 15213-1863 USA
| | - Eva Wieckowski
- Department of Surgery, Division of Surgical Oncology, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 400, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA
| | - David L Bartlett
- Department of Surgery, Division of Surgical Oncology, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 400, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA ; University of Pittsburgh Cancer Institute, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 500, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA
| | - Pawel Kalinski
- Department of Surgery, Division of Surgical Oncology, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 400, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA ; University of Pittsburgh Cancer Institute, Hillman Cancer Center, UPCI Cancer Pavilion, Suite 500, 5150 Centre Avenue, Pittsburgh, PA 15213-1863 USA ; Department of Immunology, Hillman Cancer Center, UPCI Research Pavilion, Suite 1.46, 5117 Centre Avenue, Pittsburgh, PA 15213-1863 USA ; Department of Infectious Diseases and Microbiology, Hillman Cancer Center, UPCI Research Pavilion, Suite 1.46, 5117 Centre Avenue, Pittsburgh, PA 15213-1863 USA ; Department of Bioengineering University of Pittsburgh, Hillman Cancer Center, UPCI Research Pavilion, Suite 1.46, 5117 Centre Avenue, Pittsburgh, PA 15213-1863 USA
| |
Collapse
|
14
|
Whiteside TL, Ferris RL, Szczepanski M, Tublin M, Kiss J, Johnson R, Johnson JT. Dendritic cell-based autologous tumor vaccines for head and neck squamous cell carcinoma. Head Neck 2015; 38 Suppl 1:E494-501. [PMID: 25735641 DOI: 10.1002/hed.24025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2015] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND An autologous vaccine of apoptotic tumor cells (ATCs) and dendritic cells (DCs) was administered to patients with stage III/IV head and neck squamous cell carcinoma (HNSCC) to study safety and feasibility. METHODS Autologous DCs were generated from monocytes, loaded with ATCs, and delivered intranodally. Delayed-type hypersensitivity (DTH) and immunological endpoints were measured prevaccination and postvaccination. Clinical follow-up was required. RESULTS Tumors obtained from 30 patients yielded 2 × 10(6) to 2 × 10(8) tumor cells. Only 19 of 30 (63%) were sterile. Ten of 30 patients (33%) had ≥1 × 10(7) sterile tumor cells required for vaccine production. Eight of 10 patients had positive recall DTH. Five of 10 patients were leukapheresed to generate DCs. Four of 5 patients were vaccinated. ATC-reactive T cells were detected in 3 of 4 patients. All 4 patients survived >5 years. The trial failed to enroll the projected 12 patients and was terminated. CONCLUSION This vaccine was safe and immunogenic but feasible only in patients with HNSCC with positive prevaccine DTH and ≥1 × 10(7) sterile tumor cells. All vaccinated patients were long-term disease-free survivors. © 2015 Wiley Periodicals, Inc. Head Neck 38: E494-E501, 2016.
Collapse
Affiliation(s)
- Theresa L Whiteside
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Robert L Ferris
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Miroslaw Szczepanski
- University of Pittsburgh Cancer Institute Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Mitchell Tublin
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Joseph Kiss
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Rita Johnson
- University of Pittsburgh Cancer Institute Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Jonas T Johnson
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute Hillman Cancer Center, Pittsburgh, Pennsylvania
| |
Collapse
|
15
|
Butterfield LH. Dendritic cells in cancer immunotherapy clinical trials: are we making progress? Front Immunol 2013; 4:454. [PMID: 24379816 PMCID: PMC3861778 DOI: 10.3389/fimmu.2013.00454] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 11/28/2013] [Indexed: 01/12/2023] Open
Abstract
Dendritic cells (DC) have been tested in cancer immunotherapy clinical trials for two decades. Over this time, the methods of DC culture (or manufacture) have evolved, the approaches for antigen loading have broadened, the maturation signals have varied and different sites of administration have been tested. The post-vaccination immunologic questions asked have also varied between trials and over time. In this review, I will consider multiple aspects of DC-based vaccines tested in cancer patients, including the cell culture, antigen loading, maturation, and delivery, as well as what we have learned from testing immune responses in vaccinated patients who have benefited clinically, and those who have not measurably benefited.
Collapse
Affiliation(s)
- Lisa H Butterfield
- Departments of Medicine, Surgery and Immunology, University of Pittsburgh Cancer Institute, University of Pittsburgh , Pittsburgh, PA , USA
| |
Collapse
|
16
|
Kalinski P, Muthuswamy R, Urban J. Dendritic cells in cancer immunotherapy: vaccines and combination immunotherapies. Expert Rev Vaccines 2013; 12:285-95. [PMID: 23496668 DOI: 10.1586/erv.13.22] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Dendritic cells (DCs) are specialized immunostimulatory cells involved in the induction and regulation of immune responses. The feasibility of large-scale ex vivo generation of DCs from patients' monocytes allows for therapeutic application of ex vivo-cultured DCs to bypass the dysfunction of endogenous DCs, restore immune surveillance, induce cancer regression or stabilization or delay or prevent its recurrence. While the most common paradigm of the therapeutic application of DCs reflects their use as cancer 'vaccines', additional and potentially more effective possibilities include the use of patients' autologous DCs as parts of more comprehensive therapies involving in vivo or ex vivo induction of tumor-reactive T cells and the measures to counteract systemic and local immunosuppression in tumor-bearing hosts. Ex vivo-cultured DCs can be instructed to acquire distinct functions relevant for the induction of effective cancer immunity (DC polarization), such as the induction of different effector functions or different homing properties of tumor-specific T cells (delivery of 'signal 3' and 'signal 4'). These considerations highlight the importance of the application of optimized conditions for the ex vivo culture of DCs and the potential combination of DC therapies with additional immune interventions to facilitate the entry of DC-induced T cells to tumor tissues and their local antitumor functions.
Collapse
Affiliation(s)
- Pawel Kalinski
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
| | | | | |
Collapse
|
17
|
Schilling B, Harasymczuk M, Schuler P, Egan J, Ferrone S, Whiteside TL. IRX-2, a novel immunotherapeutic, enhances functions of human dendritic cells. PLoS One 2013; 8:e47234. [PMID: 23408925 PMCID: PMC3567103 DOI: 10.1371/journal.pone.0047234] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 09/10/2012] [Indexed: 01/15/2023] Open
Abstract
Background In a recent phase II clinical trial for HNSCC patients, IRX-2, a cell-derived biologic, promoted T-cell infiltration into the tumor and prolonged overall survival. Mechanisms responsible for these IRX-2-mediated effects are unknown. We hypothesized that IRX-2 enhanced tumor antigen-(TA)-specific immunity by up-regulating functions of dendritic cells (DC). Methodology/Principal Findings Monocyte-derived DC obtained from 18 HNSCC patients and 12 healthy donors were matured using IRX-2 or a mix of TNF-α, IL-1β and IL-6 (“conv. mix”). Multicolor flow cytometry was used to study the DC phenotype and antigen processing machinery (APM) component expression. ELISPOT and cytotoxicity assays were used to evaluate tumor-reactive cytotoxic T lymphocytes (CTL). IL-12p70 and IL-10 production by DC was measured by Luminex® and DC migration toward CCL21 was tested in transwell migration assays. IRX-2-matured DC functions were compared with those of conv. mix-matured DC. IRX-2-matured DC expressed higher levels (p<0.05) of CD11c, CD40, CCR7 as well as LMP2, TAP1, TAP2 and tapasin than conv. mix-matured DC. IRX-2-matured DC migrated significantly better towards CCL21, produced more IL-12p70 and had a higher IL12p70/IL-10 ratio than conv. mix-matured DC (p<0.05 for all). IRX-2-matured DC carried a higher density of tumor antigen-derived peptides, and CTL primed with these DC mediated higher cytotoxicity against tumor targets (p<0.05) compared to the conv. mix-matured DC. Conclusion Excellent ability of IRX-2 to induce ex vivo DC maturation in HNSCC patients explains, in part, its clinical benefits and emphasizes its utility in ex vivo maturation of DC generated for therapy.
Collapse
Affiliation(s)
- Bastian Schilling
- University of Pittsburgh, Department of Pathology and University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Malgorzata Harasymczuk
- University of Pittsburgh, Department of Pathology and University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Patrick Schuler
- University of Pittsburgh, Department of Pathology and University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - James Egan
- IRX Therapeutic Inc., Farmingdale, New York, United States of America
| | - Soldano Ferrone
- University of Pittsburgh, Department of Pathology and University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Theresa L. Whiteside
- University of Pittsburgh, Department of Pathology and University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
18
|
Comparison of clinical grade type 1 polarized and standard matured dendritic cells for cancer immunotherapy. Vaccine 2013. [DOI: 10.1016/j.vaccine.2012.11.053] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
19
|
Berk E, Muthuswamy R, Kalinski P. Lymphocyte-polarized dendritic cells are highly effective in inducing tumor-specific CTLs. Vaccine 2012; 30:6216-24. [PMID: 22561311 DOI: 10.1016/j.vaccine.2012.04.077] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 04/19/2012] [Accepted: 04/21/2012] [Indexed: 10/28/2022]
Abstract
High activity of dendritic cells (DCs) in inducing cytotoxic T cells (CTLs) led to their application as therapeutic cancer vaccines. The ability of DCs to produce IL-12p70 is one of the key requirements for effective CTL induction and a predictive marker of their therapeutic efficacy in vivo. We have previously reported that defined cocktails of cytokines, involving TNFα and IFNγ, induce mature type-1 polarized DCs (DC1s) which produce strongly elevated levels of IL-12 and CXCL10/IP10 upon CD40 ligation compared to "standard" PGE₂-matured DCs (sDCs; matured with IL-1β, IL-6, TNFα, and PGE₂) and show higher CTL-inducing activity. Guided by our observations that DC1s can be induced by TNFα- and IFNγ-producing CD8⁺ T cells, we have tested the feasibility of using lymphocytes to generate DC1s in a clinically-compatible process, to limit the need for clinical-grade recombinant cytokines and the associated costs. CD3/CD28 activation of bulk lymphocytes expanded them and primed them for effective production of IFNγ and TNFα following restimulation. Restimulated lymphocytes, or their culture supernatants, enhanced the maturation status of immature (i)DCs, elevating their expression of CD80, CD83 and CCR7, and the ability to produce IL-12p70 and CXCL10 upon subsequent CD40 ligation. The "lymphocyte-matured" DC1s showed elevated migration in response to the lymph-node-directing chemokine, CCL21, when compared to iDCs. When loaded with antigenic peptides, supernatant-matured DCs induced much high levels of CTLs recognizing tumor-associated antigenic epitope, than PGE₂-matured DCs from the same donors. These results demonstrate the feasibility of generation of polarized DC1s using autologous lymphocytes.
Collapse
Affiliation(s)
- Erik Berk
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | |
Collapse
|
20
|
Xu H, Cao X. Dendritic cell vaccines in cancer immunotherapy: from biology to translational medicine. Front Med 2012; 5:323-32. [PMID: 22198743 DOI: 10.1007/s11684-011-0172-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Hongmei Xu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China.
| | | |
Collapse
|
21
|
Abstract
Immunotherapy with dendritic cells (DCs), which have been manipulated ex vivo to become immunogenic or tolerogenic, has been tested in clinical trials for disease therapy. DCs are sentinels of the immune system, which after exposure to antigenic or inflammatory signals and crosstalk with effector CD4(+) T cells express high levels of costimulatory molecules and cytokines. Upregulation of either costimulatory molecules or cytokines promotes immunologic DCs, whereas their downregulation generates tolerogenic DCs (TDCs), which induce T regulatory cells (Tregs) and a state of tolerance. Immunogenic DCs are used for the therapy of infectious diseases such as HIV-1 and cancer, whereas tolerogenic DCs are used in treating various autoimmune diseases and in transplantation. DC vaccination is still at an early stage, and improvements are mainly needed in quality control of monitoring assays to generate clinical-grade DC products and to assess the effect of DC vaccination in future clinical trials. Here, we review the recent work in DC generation and monitoring approaches for DC-based trials with immunogenic or tolerogenic DCs.
Collapse
|
22
|
Cryopreservation of adenovirus-transfected dendritic cells (DCs) for clinical use. Int Immunopharmacol 2012; 13:61-8. [PMID: 22465385 DOI: 10.1016/j.intimp.2012.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 03/15/2012] [Indexed: 11/22/2022]
Abstract
In this study, we examined the effects of cryoprotectant, freezing and thawing, and adenovirus (Adv) transduction on the viability, transgene expression, phenotype, and function of human dendritic cells (DCs). DCs were differentiated from cultured peripheral blood (PB) monocytes following Elutra isolation using granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4) for 6 days and then transduced using an Adv vector with an IL-12 transgene. Fresh, cryopreserved, and thawed transduced immature DCs were examined for their: 1) cellular concentration and viability; 2) antigenicity using an allogeneic mixed lymphocyte reaction (MLR); 3) phenotype (HLA-DR and CD11c) and activation (CD83); and 4) transgene expression based on IL-12 secretion. Stability studies revealed that transduced DCs could be held in cryoprotectant for as long as 75 min at 2-8°C prior to freezing with little effect on their viability and cellularity. Further, cryopreservation, storage, and thawing reduced the viability of the transduced DCs by an average of 7.7%; and had no significant impact on DC phenotype and activation. In summary, cryopreservation, storage, and thawing had no significant effect on DC viability, function, and transgene expression by Adv-transduced DCs.
Collapse
|
23
|
Reconsidering the Paradigm of Cancer Immunotherapy by Computationally Aided Real-time Personalization. Cancer Res 2012; 72:2218-27. [DOI: 10.1158/0008-5472.can-11-4166] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
24
|
Chiang CLL, Maier DA, Kandalaft LE, Brennan AL, Lanitis E, Ye Q, Levine BL, Czerniecki BJ, Powell DJ, Coukos G. Optimizing parameters for clinical-scale production of high IL-12 secreting dendritic cells pulsed with oxidized whole tumor cell lysate. J Transl Med 2011; 9:198. [PMID: 22082029 PMCID: PMC3283529 DOI: 10.1186/1479-5876-9-198] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 11/14/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dendritic cells (DCs) are the most potent antigen-presenting cell population for activating tumor-specific T cells. Due to the wide range of methods for generating DCs, there is no common protocol or defined set of criteria to validate the immunogenicity and function of DC vaccines. METHODS Monocyte-derived DCs were generated during 4 days of culture with recombinant granulocyte-macrophage colony stimulating factor and interleukin-4, and pulsed with tumor lysate produced by hypochlorous acid oxidation of tumor cells. Different culture parameters for clinical-scale DC preparation were investigated, including: 1) culture media; 2) culture surface; 3) duration of activating DCs with lipopolysaccharide (LPS) and interferon (IFN)-gamma; 4) method of DC harvest; and 5) cryomedia and final DC product formulation. RESULTS DCs cultured in CellGenix DC media containing 2% human AB serum expressed higher levels of maturation markers following lysate-loading and maturation compared to culturing with serum-free CellGenix DC media or AIM-V media, or 2% AB serum supplemented AIM-V media. Nunclon™Δ surface, but not Corning(®) tissue-culture treated surface and Corning(®) ultra-low attachment surface, were suitable for generating an optimal DC phenotype. Recombinant trypsin resulted in reduced major histocompatibility complex (MHC) Class I and II expression on mature lysate-loaded DCs, however presentation of MHC Class I peptides by DCs was not impaired and cell viability was higher compared to cell scraping. Preservation of DCs with an infusible cryomedia containing Plasma-Lyte A, dextrose, sodium chloride injection, human serum albumin, and DMSO yielded higher cell viability compared to using human AB serum containing 10% DMSO. Finally, activating DCs for 16 hours with LPS and IFN-γ stimulated robust mixed leukocyte reactions (MLRs), and high IL-12p70 production in vitro that continued for 24 hours after the cryopreserved DCs were thawed and replated in fresh media. CONCLUSIONS This study examined criteria including DC phenotype, viability, IL-12p70 production and the ability to stimulate MLR as metrics of whole oxidized tumor lysate-pulsed DC immunogenicity and functionality. Development and optimization of this unique method is now being tested in a clinical trial of autologous oxidized tumor lysate-pulsed DC in clinical-scale in recurrent ovarian, primary peritoneal or fallopian tube cancer (NCT01132014).
Collapse
Affiliation(s)
- Cheryl L-L Chiang
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Dendritic cells (DCs) are the most powerful immunostimulatory cells specialized in the induction and regulation of immune responses. Their properties and the feasibility of their large-scale ex vivo generation led to the application of ex vivo-educated DCs to bypass the dysfunction of endogenous DCs in cancer patients and to induce therapeutic anti-cancer immunity. While multiple paradigms of therapeutic application of DCs reflect their consideration as cancer "vaccines", numerous features of DC-based vaccination resemble those of autologous transplants, resulting in challenges and opportunities that distinguish them from classical vaccines. In addition to the functional heterogeneity of DC subsets and plasticity of the individual DC types, the unique features of DCs are the kinetic character of their function, limited functional stability, and the possibility to imprint in maturing DCs distinct functions relevant for the induction of effective cancer immunity, such as the induction of different effector functions or different homing properties of tumor-specific T cells (delivery of "signal 3" and "signal 4"). These considerations highlight the importance of the application of optimized, potentially patient-specific conditions of ex vivo culture of DCs and their delivery, with the logistic and regulatory implications shared with transplantation and other surgical procedures.
Collapse
|
26
|
Butterfield LH, Potter DM, Kirkwood JM. Multiplex serum biomarker assessments: technical and biostatistical issues. J Transl Med 2011; 9:173. [PMID: 21989127 PMCID: PMC3200183 DOI: 10.1186/1479-5876-9-173] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 10/11/2011] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Identification of predictive and prognostic biomarkers for patients with disease and undergoing different therapeutic options is a very active area of investigation. Many of these studies seek biomarkers among circulating proteins accessed in blood. Many levels of standardization in materials and procedures have been identified which can impact the resulting data. METHODS Here, we have observed unexpected variability in levels of commonly tested analytes in serum which were processed and stored under standardized conditions. We have identified apparent changes in cytokine, chemokine and growth factor levels detected by multiplex Luminex assay in melanoma patient and healthy donor serum samples, over storage time at -80°C. Controls included Luminex kit standards, multiplexed cytokine standards and WHO cytokine controls. Data were analyzed by Wilcoxon rank-sum testing and Spearman's test for correlations. RESULTS The interpretation of these changes is confounded by lot-to-lot kit standard curve reagent changes made by a single manufacturer of Luminex kits. CONCLUSIONS This study identifies previously unknown sources of variation in a commonly used biomarker assay, and suggests additional levels of controls needed for identification of true changes in circulating protein levels.
Collapse
Affiliation(s)
- Lisa H Butterfield
- University of Pittsburgh Cancer Institute, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- University of Pittsburgh School of Medicine, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- Department of Surgery, University of Pittsburgh, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
| | - Douglas M Potter
- University of Pittsburgh Cancer Institute, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- University of Pittsburgh School of Medicine, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- Depament of Biostatistics, University of Pittsburgh, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
| | - John M Kirkwood
- University of Pittsburgh Cancer Institute, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- University of Pittsburgh School of Medicine, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
| |
Collapse
|
27
|
Butterfield LH, Palucka AK, Britten CM, Dhodapkar MV, Håkansson L, Janetzki S, Kawakami Y, Kleen TO, Lee PP, Maccalli C, Maecker HT, Maino VC, Maio M, Malyguine A, Masucci G, Pawelec G, Potter DM, Rivoltini L, Salazar LG, Schendel DJ, Slingluff CL, Song W, Stroncek DF, Tahara H, Thurin M, Trinchieri G, van Der Burg SH, Whiteside TL, Wigginton JM, Marincola F, Khleif S, Fox BA, Disis ML. Recommendations from the iSBTc-SITC/FDA/NCI Workshop on Immunotherapy Biomarkers. Clin Cancer Res 2011; 17:3064-76. [PMID: 21558394 DOI: 10.1158/1078-0432.ccr-10-2234] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To facilitate development of innovative immunotherapy approaches, especially for treatment concepts exploiting the potential benefits of personalized therapy, there is a need to develop and validate tools to identify patients who can benefit from immunotherapy. Despite substantial effort, we do not yet know which parameters of antitumor immunity to measure and which assays are optimal for those measurements. EXPERIMENTAL DESIGN The iSBTc-SITC (International Society for Biological Therapy of Cancer-Society for Immunotherapy of Cancer), FDA (Food and Drug Administration), and NCI (National Cancer Institute) partnered to address these issues for immunotherapy of cancer. Here, we review the major challenges, give examples of approaches and solutions, and present our recommendations. RESULTS AND CONCLUSIONS Although specific immune parameters and assays are not yet validated, we recommend following standardized (accurate, precise, and reproducible) protocols and use of functional assays for the primary immunologic readouts of a trial; consideration of central laboratories for immune monitoring of large, multi-institutional trials; and standardized testing of several phenotypic and functional potential potency assays specific to any cellular product. When reporting results, the full QA (quality assessment)/QC (quality control) should be conducted and selected examples of truly representative raw data and assay performance characteristics should be included. Finally, to promote broader analysis of multiple aspects of immunity, and gather data on variability, we recommend that in addition to cells and serum, RNA and DNA samples be banked (under standardized conditions) for later testing. We also recommend that sufficient blood be drawn to allow for planned testing of the primary hypothesis being addressed in the trial, and that additional baseline and posttreatment blood is banked for testing novel hypotheses (or generating new hypotheses) that arise in the field.
Collapse
Affiliation(s)
- Lisa H Butterfield
- Department of Medicine, University of Pittsburgh, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Whiteside TL, Griffin DL, Stanson J, Gooding W, McKenna D, Sumstad D, Kadidlo D, Gee A, Durett A, Lindblad R, Wood D, Styers D. Shipping of therapeutic somatic cell products. Cytotherapy 2011; 13:201-13. [PMID: 20795760 PMCID: PMC7982143 DOI: 10.3109/14653249.2010.506507] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Shipment of therapeutic somatic cells between a current good manufacturing practice (cGMP) facility and a clinic or between different cGMP facilities requires validated standard operating procedures (SOP). Under National Heart Lung & Blood Institute (NHLBI) sponsorship, the Production Assistance for Cellular Therapies (PACT) group conducted a validation study for the shipping SOP it has created, including shipments of cryopreserved somatic cells, fresh peripheral blood specimens and apheresis products. METHODS Comparisons of pre- and post-shipped cells and cell products at the three participating facilities included measurements of viability, phenotypic profiles and cellular functions. The data were analyzed at the University of Pittsburgh Biostatistics Facility. RESULTS No consistent shipping effects on cell viability, phenotype or functions were detected for cryopreserved and shipped peripheral blood mononuclear cells (PBMC), monocytes, immature dendritic cells (iDC), NK-92 or cytotoxic T cells (CTL). Cryopreserved mesenchymal stromal cells (MSC) had a significantly decreased viability after shipment, but this effect was in part because of inter-laboratory variability in the viable cell counts. Shipments of fresh peripheral blood and apheresis products for the generation of CTL and dendritic cells (DC), respectively, had no significant effects on cell product quality. MSC were successfully generated from fresh bone marrow samples shipped overnight. CONCLUSIONS This validation study provides a useful set of data for guiding shipments of therapeutic somatic cells in multi-institutional clinical trials.
Collapse
|
29
|
Barth RJ, Fisher DA, Wallace PK, Channon JY, Noelle RJ, Gui J, Ernstoff MS. A randomized trial of ex vivo CD40L activation of a dendritic cell vaccine in colorectal cancer patients: tumor-specific immune responses are associated with improved survival. Clin Cancer Res 2010; 16:5548-56. [PMID: 20884622 DOI: 10.1158/1078-0432.ccr-10-2138] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE To determine whether an autologous dendritic cell (DC) vaccine could induce antitumor immune responses in patients after resection of colorectal cancer metastases and whether these responses could be enhanced by activating DCs with CD40L. EXPERIMENTAL DESIGN Twenty-six patients who had undergone resection of colorectal metastases were treated with intranodal injections of an autologous tumor lysate- and control protein [keyhole limpet hemocyanin (KLH)]-pulsed DC vaccine. Patients were randomized to receive DCs that had been either activated or not activated with CD40L. All patients were followed for a minimum of 5.5 years. RESULTS Immunization induced an autologous tumor-specific T-cell proliferative or IFNγ enzyme-linked immunospot response in 15 of 24 assessable patients (63%) and a tumor-specific DTH response in 61%. Patients with evidence of a vaccine-induced, tumor-specific T-cell proliferative or IFNγ response 1 week after vaccination had a markedly better recurrence-free survival (RFS) at 5 years (63% versus 18%, P = 0.037) than nonresponders. In contrast, no association was observed between induction of KLH-specific immune responses and RFS. CD40L maturation induced CD86 and CD83 expression on DCs but had no effect on immune responses or RFS. CONCLUSION Adjuvant treatment of patients after resection of colorectal metastases with an autologous tumor lysate-pulsed, DC vaccine-induced, tumor-specific immune responses in a high proportion of patients. There was an association between induction of tumor-specific immune responses and RFS. Activation of this DC vaccine with CD40L did not lead to increased immune responses.
Collapse
Affiliation(s)
- Richard J Barth
- Department of Surgery, Section of General Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire 03756, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Evaluation of 3 clinical dendritic cell maturation protocols containing lipopolysaccharide and interferon-gamma. J Immunother 2009; 32:399-407. [PMID: 19342965 DOI: 10.1097/cji.0b013e31819e1773] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) are important adjuvants for cancer vaccines. Immature dendritic cells (iDCs) are often produced by the stimulation of peripheral blood monocytes with interleukin (IL)-4 and granulocyte macrophage-colony stimulating factor. For many applications iDCs are treated with cytokines or inflammatory signals to produce mature DCs (mDCs). iDCs are often treated ex vivo with lipopolysaccharide (LPS) and interferon (IFN)-gamma to produce mDCs for clinical therapy. The purpose of this study was to determine if the DC maturation cocktail LPS plus IFN-gamma could be improved by the addition of 2 other DC maturation agents IL-1beta and tumor necrosis factor (TNF)-alpha. Peripheral blood mononuclear cells were collected from 6 healthy subjects. Monocytes were isolated from the peripheral blood mononuclear cell concentrates by elutriation and were incubated for 3 days with granulocyte macrophage-colony stimulating factor and IL-4 to produce iDCs. iDCs from each subject were divided into 3 and were incubated for 24 hours with LPS plus IFN-gamma; LPS, IFN-gamma, plus IL-1beta; or LPS, IFN-gamma, IL-1beta, plus TNF-alpha to produce mDCs. The DCs were compared by measuring the expression of costimulator and antigen presenting molecules (CD80, CD83, CD86, and human leukocyte antigen-DR) by flow cytometry, cytokine production (IL-12p70 and IL-10) by enzyme-linked immunosorbent assay and global gene expression using an oligonucleotide microarray. There were no differences in the expression of costimulatory molecules, human leukocyte antigen-DR and CCR7 and production of IL-12p70 among the mDCs produced with the 3 cocktails. Global gene expression analysis found that the expression of 9576 genes differed between the iDCs and mDCs, but the expression of only 13 differed among the 3 different groups of mDCs. There was no benefit of adding IL-1beta and TNF-alpha to LPS and IFN-gamma to produce mDCs.
Collapse
|
31
|
Tahara H, Sato M, Thurin M, Wang E, Butterfield LH, Disis ML, Fox BA, Lee PP, Khleif SN, Wigginton JM, Ambs S, Akutsu Y, Chaussabel D, Doki Y, Eremin O, Fridman WH, Hirohashi Y, Imai K, Jacobson J, Jinushi M, Kanamoto A, Kashani-Sabet M, Kato K, Kawakami Y, Kirkwood JM, Kleen TO, Lehmann PV, Liotta L, Lotze MT, Maio M, Malyguine A, Masucci G, Matsubara H, Mayrand-Chung S, Nakamura K, Nishikawa H, Palucka AK, Petricoin EF, Pos Z, Ribas A, Rivoltini L, Sato N, Shiku H, Slingluff CL, Streicher H, Stroncek DF, Takeuchi H, Toyota M, Wada H, Wu X, Wulfkuhle J, Yaguchi T, Zeskind B, Zhao Y, Zocca MB, Marincola FM. Emerging concepts in biomarker discovery; the US-Japan Workshop on Immunological Molecular Markers in Oncology. J Transl Med 2009; 7:45. [PMID: 19534815 PMCID: PMC2724494 DOI: 10.1186/1479-5876-7-45] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Accepted: 06/17/2009] [Indexed: 02/08/2023] Open
Abstract
Supported by the Office of International Affairs, National Cancer Institute (NCI), the "US-Japan Workshop on Immunological Biomarkers in Oncology" was held in March 2009. The workshop was related to a task force launched by the International Society for the Biological Therapy of Cancer (iSBTc) and the United States Food and Drug Administration (FDA) to identify strategies for biomarker discovery and validation in the field of biotherapy. The effort will culminate on October 28th 2009 in the "iSBTc-FDA-NCI Workshop on Prognostic and Predictive Immunologic Biomarkers in Cancer", which will be held in Washington DC in association with the Annual Meeting. The purposes of the US-Japan workshop were a) to discuss novel approaches to enhance the discovery of predictive and/or prognostic markers in cancer immunotherapy; b) to define the state of the science in biomarker discovery and validation. The participation of Japanese and US scientists provided the opportunity to identify shared or discordant themes across the distinct immune genetic background and the diverse prevalence of disease between the two Nations. Converging concepts were identified: enhanced knowledge of interferon-related pathways was found to be central to the understanding of immune-mediated tissue-specific destruction (TSD) of which tumor rejection is a representative facet. Although the expression of interferon-stimulated genes (ISGs) likely mediates the inflammatory process leading to tumor rejection, it is insufficient by itself and the associated mechanisms need to be identified. It is likely that adaptive immune responses play a broader role in tumor rejection than those strictly related to their antigen-specificity; likely, their primary role is to trigger an acute and tissue-specific inflammatory response at the tumor site that leads to rejection upon recruitment of additional innate and adaptive immune mechanisms. Other candidate systemic and/or tissue-specific biomarkers were recognized that might be added to the list of known entities applicable in immunotherapy trials. The need for a systematic approach to biomarker discovery that takes advantage of powerful high-throughput technologies was recognized; it was clear from the current state of the science that immunotherapy is still in a discovery phase and only a few of the current biomarkers warrant extensive validation. It was, finally, clear that, while current technologies have almost limitless potential, inadequate study design, limited standardization and cross-validation among laboratories and suboptimal comparability of data remain major road blocks. The institution of an interactive consortium for high throughput molecular monitoring of clinical trials with voluntary participation might provide cost-effective solutions.
Collapse
Affiliation(s)
- Hideaki Tahara
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Marimo Sato
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Magdalena Thurin
- Cancer Diagnosis Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, Maryland, 20852, USA
| | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Center for Human Immunology (CHI), NIH, Bethesda, Maryland, 20892, USA
| | - Lisa H Butterfield
- Departments of Medicine, Surgery and Immunology, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, 15213, USA
| | - Mary L Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, Washington, 98195, USA
| | - Bernard A Fox
- Earle A Chiles Research Institute, Robert W Franz Research Center, Providence Portland Medical Center, and Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, 97213, USA
| | - Peter P Lee
- Department of Medicine, Division of Hematology, Stanford University, Stanford, California, 94305, USA
| | - Samir N Khleif
- Cancer Vaccine Section, NCI, NIH, Bethesda, Maryland, 20892, USA
| | - Jon M Wigginton
- Discovery Medicine-Oncology, Bristol-Myers Squibb Inc., Princeton, New Jersey, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center of Cancer Research, NCI, NIH, Bethesda, Maryland, 20892, USA
| | - Yasunori Akutsu
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Damien Chaussabel
- Baylor Institute for Immunology Research and Baylor Research Institute, Dallas, Texas, 75204, USA
| | - Yuichiro Doki
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Oleg Eremin
- Section of Surgery, Biomedical Research Unit, Nottingham Digestive Disease Centre, University of Nottingham, NG7 2UH, UK
| | - Wolf Hervé Fridman
- Centre de la Reserche des Cordeliers, INSERM, Paris Descarte University, 75270 Paris, France
| | | | - Kohzoh Imai
- Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - James Jacobson
- Cancer Diagnosis Program, National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, Maryland, 20852, USA
| | - Masahisa Jinushi
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Akira Kanamoto
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | - Kazunori Kato
- Department of Molecular Medicine, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - John M Kirkwood
- Departments of Medicine, Surgery and Immunology, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, 15213, USA
| | - Thomas O Kleen
- Cellular Technology Ltd, Shaker Heights, Ohio, 44122, USA
| | - Paul V Lehmann
- Cellular Technology Ltd, Shaker Heights, Ohio, 44122, USA
| | - Lance Liotta
- Department of Molecular Pathology and Microbiology, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia, 10900, USA
| | - Michael T Lotze
- Illman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213, USA
| | - Michele Maio
- Medical Oncology and Immunotherapy, Department. of Oncology, University, Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
- Cancer Bioimmunotherapy Unit, Department of Medical Oncology, Centro di Riferimento Oncologico, IRCCS, Aviano, 53100, Italy
| | - Anatoli Malyguine
- Laboratory of Cell Mediated Immunity, SAIC-Frederick, Inc. NCI-Frederick, Frederick, Maryland, 21702, USA
| | - Giuseppe Masucci
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, 171 76, Sweden
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shawmarie Mayrand-Chung
- The Biomarkers Consortium (BC), Public-Private Partnership Program, Office of the Director, NIH, Bethesda, Maryland, 20892, USA
| | - Kiminori Nakamura
- Department of Molecular Medicine, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Hiroyoshi Nishikawa
- Department of Cancer Vaccine, Department of Immuno-gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - A Karolina Palucka
- Baylor Institute for Immunology Research and Baylor Research Institute, Dallas, Texas, 75204, USA
| | - Emanuel F Petricoin
- Department of Molecular Pathology and Microbiology, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia, 10900, USA
| | - Zoltan Pos
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Center for Human Immunology (CHI), NIH, Bethesda, Maryland, 20892, USA
| | - Antoni Ribas
- Department of Medicine, Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, 90095, USA
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, IRCCS Foundation, Istituto Nazionale Tumori, Milan, 20100, Italy
| | - Noriyuki Sato
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroshi Shiku
- Department of Cancer Vaccine, Department of Immuno-gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Craig L Slingluff
- Department of Surgery, Division of Surgical Oncology, University of Virginia School of Medicine, Charlottesville, Virginia, 22908, USA
| | - Howard Streicher
- Cancer Therapy Evaluation Program, DCTD, NCI, NIH, Rockville, Maryland, 20892, USA
| | - David F Stroncek
- Cell Therapy Section (CTS), Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland, 20892, USA
| | - Hiroya Takeuchi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Minoru Toyota
- Department of Biochemistry, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Hisashi Wada
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Xifeng Wu
- Department of Epidemiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Julia Wulfkuhle
- Department of Molecular Pathology and Microbiology, Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia, 10900, USA
| | - Tomonori Yaguchi
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | | | - Yingdong Zhao
- Biometric Research Branch, NCI, NIH, Bethesda, Maryland, 20892, USA
| | | | - Francesco M Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Center for Human Immunology (CHI), NIH, Bethesda, Maryland, 20892, USA
| |
Collapse
|
32
|
Tcherepanova I, Starr A, Lackford B, Adams MD, Routy JP, Boulassel MR, Calderhead D, Healey D, Nicolette C. The immunosuppressive properties of the HIV Vpr protein are linked to a single highly conserved residue, R90. PLoS One 2009; 4:e5853. [PMID: 19516896 PMCID: PMC2689350 DOI: 10.1371/journal.pone.0005853] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Accepted: 05/05/2009] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND A hallmark of AIDS progression is a switch of cytokines from Th1 to Th2 in the plasma of patients. IL-12, a critical Th1 cytokine secreted by antigen presenting cells (APCs) is suppressed by Vpr, implicating it as an important virulence factor. We hypothesize that Vpr protein packaged in the virion may be required for disabling APCs of the first infected mucosal tissues. Consistent with this idea are reports that defects in the C-terminus of Vpr are associated with long-term non-progression. PRINCIPAL FINDINGS Vpr RNA amplified from various sources was electroporated into monocyte-derived DC and IL-12 levels in supernatants were analyzed. The analysis of previously reported C-terminal Vpr mutations demonstrate that they do not alleviate the block of IL-12 secretion. However, a novel single conservative amino acid substitution, R90K, reverses the IL-12 suppression. Analysis of 1226 Vpr protein sequences demonstrated arginine (R) present at position 90 in 98.8%, with other substitutions at low frequency. Furthermore, none of sequences report lysine (K) in position 90. Vpr clones harboring the reported substitutions in position 90 were studied for their ability to suppress IL-12. Our data demonstrates that none of tested substitutions other than K relieve IL-12 suppression. This suggests a natural selection for sequences which suppress IL-12 secretion by DC and against mutations which relieve such suppression. Further analyses demonstrated that the R90K, as well as deletion of the C-terminus, directs the Vpr protein for rapid degradation. CONCLUSION This study supports Vpr as an HIV virulence factor during HIV infection and for the first time provides a link between evolutionary conservation of Vpr and its ability to suppress IL-12 secretion by DC. DC activated in the presence of Vpr would be defective in the production of IL-12, thus contributing to the prevailing Th2 cytokine profile associated with progressive HIV disease. These findings should be considered in the design of future immunotherapies that incorporate Vpr as an antigen.
Collapse
Affiliation(s)
- Irina Tcherepanova
- Research and Development Department, Argos Therapeutics Inc, Durham, NC, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Butterfield LH, Disis ML, Fox BA, Lee PP, Khleif SN, Thurin M, Trinchieri G, Wang E, Wigginton J, Chaussabel D, Coukos G, Dhodapkar M, Håkansson L, Janetzki S, Kleen TO, Kirkwood JM, Maccalli C, Maecker H, Maio M, Malyguine A, Masucci G, Palucka AK, Potter DM, Ribas A, Rivoltini L, Schendel D, Seliger B, Selvan S, Slingluff CL, Stroncek DF, Streicher H, Wu X, Zeskind B, Zhao Y, Zocca MB, Zwierzina H, Marincola FM. A systematic approach to biomarker discovery; preamble to "the iSBTc-FDA taskforce on immunotherapy biomarkers". J Transl Med 2008; 6:81. [PMID: 19105846 PMCID: PMC2630944 DOI: 10.1186/1479-5876-6-81] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Accepted: 12/23/2008] [Indexed: 12/23/2022] Open
Abstract
The International Society for the Biological Therapy of Cancer (iSBTc) has initiated in collaboration with the United States Food and Drug Administration (FDA) a programmatic look at innovative avenues for the identification of relevant parameters to assist clinical and basic scientists who study the natural course of host/tumor interactions or their response to immune manipulation. The task force has two primary goals: 1) identify best practices of standardized and validated immune monitoring procedures and assays to promote inter-trial comparisons and 2) develop strategies for the identification of novel biomarkers that may enhance our understating of principles governing human cancer immune biology and, consequently, implement their clinical application. Two working groups were created that will report the developed best practices at an NCI/FDA/iSBTc sponsored workshop tied to the annual meeting of the iSBTc to be held in Washington DC in the Fall of 2009. This foreword provides an overview of the task force and invites feedback from readers that might be incorporated in the discussions and in the final document.
Collapse
Affiliation(s)
- Lisa H Butterfield
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, 15213, USA
| | - Mary L Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, Washington, 98195, USA
| | - Bernard A Fox
- Earle A Chiles Research Institute, Providence Portland Medical Center, Portland, Oregon, 97213, USA
- Department of Molecular Biology, OHSU Cancer Institute, Oregon Health and Science University, Portland, Oregon, 97213, USA
| | - Peter P Lee
- Department of Medicine, Division of Hematology, Stanford University, Stanford, California, 94305, USA
| | - Samir N Khleif
- Cancer Vaccine Section, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Magdalena Thurin
- Cancer Diagnosis Program, NCI, NIH, Rockville, Maryland, 20852, USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, NCI, NIH, Frederick, Maryland, 21702, USA
| | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Center for Human Immunology, National Institutes of Health, Bethesda, MD, USA
| | - Jon Wigginton
- Bristol Myers-Squibb, Princeton, New Jersey, 08540, USA
| | - Damien Chaussabel
- Baylor Institute for Immunology Research and Baylor Research Institute, Dallas, Texas, 75204, USA
| | - George Coukos
- Center for Research on the Early Detection and Cure of Ovarian Cancer, University of Pennsylvania, Philadelphia 19104, USA
| | - Madhav Dhodapkar
- Department of Hematology, Yale University, New Haven, Connecticut 06510, USA
| | - Leif Håkansson
- Division of Clinical Tumor Immunology, University of Lund, 581 85, Sweden
| | | | - Thomas O Kleen
- Cellular Technology Limited, Shaker Heights, Ohio, 44122, USA
| | - John M Kirkwood
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, 15213, USA
| | - Cristina Maccalli
- Unit of Immuno-Biotherapy of Solid Tumors, Department of Molecular Oncology, San Raffaele Scientific Institute DIBIT, Milan, 20132, Italy
| | - Holden Maecker
- Baylor Institute for Immunology Research, Dallas, 75204, Texas, USA
| | - Michele Maio
- Medical Oncology and Immunotherapy, Department. of Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
- Cancer Bioimmunotherapy Unit, Department of Medical Oncology, Centro di Riferimento Oncologico, IRCCS, Aviano, 53100, Italy
| | - Anatoli Malyguine
- Laboratory of Cell Mediated Immunity, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD, 21702, USA
| | - Giuseppe Masucci
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, 171 76, Sweden
| | - A Karolina Palucka
- Baylor Institute for Immunology Research and Baylor Research Institute, Dallas, Texas, 75204, USA
| | - Douglas M Potter
- Biostatistics Department, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213, USA
| | - Antoni Ribas
- Department of Medicine, Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, 90095, USA
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, IRCCS Foundation, Istituto Nazionale Tumori, Milan, 20100, Italy
| | - Dolores Schendel
- Institute of Molecular Immunology, and Clinical Cooperation Group "Immune Monitoring" Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, 81377, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin-Luther University, Halle Wittenberg, Halle (Saale), 06112, Germany
| | | | - Craig L Slingluff
- Department of Surgery, Division of Surgical Oncology, University of Virginia School of Medicine, Charlottesville, Virginia, 22908, USA
| | - David F Stroncek
- Cell Therapy Section, Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland, 20892, USA
| | - Howard Streicher
- Cancer Therapy Evaluation Program, NCI, Bethesda, Maryland, 20852 USA
| | - Xifeng Wu
- Department of Epidemiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | | | - Yingdong Zhao
- Biometrics Research Branch, NCI, NIH, Bethesda, Maryland, 20852, USA
| | | | - Heinz Zwierzina
- Department of Internal Medicine, Innsbruck Medical University, Innsbruck, 6020, Austria
| | - Francesco M Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Center for Human Immunology, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
34
|
Tcherepanova IY, Adams MD, Feng X, Hinohara A, Horvatinovich J, Calderhead D, Healey D, Nicolette CA. Ectopic expression of a truncated CD40L protein from synthetic post-transcriptionally capped RNA in dendritic cells induces high levels of IL-12 secretion. BMC Mol Biol 2008; 9:90. [PMID: 18928538 PMCID: PMC2576345 DOI: 10.1186/1471-2199-9-90] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Accepted: 10/17/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND RNA transfection into dendritic cells (DCs) is widely used to achieve antigen expression as well as to modify DC properties. CD40L is expressed by activated T cells and interacts with CD40 receptors expressed on the surface of the DCs leading to Th1 polarization. Previous studies demonstrated that ectopic CD40L expression via DNA transfection into DCs can activate the CD40 receptor signal transduction cascade. In contrast to previous reports, this study demonstrates that the same effect can be achieved when RNA encoding CD40L is electroporated into DCs as evidenced by secretion of IL-12. To achieve higher levels of IL-12 secretion, a systematic approach involving modification of coding and noncoding regions was implemented to optimize protein expression in the DCs for the purpose of increasing IL-12 secretion. RESULTS Site-directed mutagenesis of each of the first five in-frame methionine codons in the CD40L coding sequence demonstrated that DCs expressing a truncated CD40L protein initiated from the second methionine codon secreted the highest levels of IL-12. In addition, a post-transcriptional method of capping was utilized for final modification of the CD40L RNA. This method enzymatically creates a type I cap structure identical to that found in most eukaryotic mRNAs, in contrast to the type 0 cap incorporated using the conventional co-transcriptional capping reaction. CONCLUSION The combination of knocking out the first initiation methionine and post-transcriptional capping of the CD40L RNA allowed for approximately a one log increase in IL-12 levels by the transfected DCs. We believe this is a first report describing improved protein expression of post-transcriptionally capped RNA in DCs. The post-transcriptional capping which allows generation of a type I cap may have broad utility for optimization of protein expression from RNA in DCs and other cell types.
Collapse
|