1
|
Foley CR, Swan SL, Swartz MA. Engineering Challenges and Opportunities in Autologous Cellular Cancer Immunotherapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:188-198. [PMID: 38166251 PMCID: PMC11155266 DOI: 10.4049/jimmunol.2300642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/18/2023] [Indexed: 01/04/2024]
Abstract
The use of a patient's own immune or tumor cells, manipulated ex vivo, enables Ag- or patient-specific immunotherapy. Despite some clinical successes, there remain significant barriers to efficacy, broad patient population applicability, and safety. Immunotherapies that target specific tumor Ags, such as chimeric Ag receptor T cells and some dendritic cell vaccines, can mount robust immune responses against immunodominant Ags, but evolving tumor heterogeneity and antigenic downregulation can drive resistance. In contrast, whole tumor cell vaccines and tumor lysate-loaded dendritic cell vaccines target the patient's unique tumor antigenic repertoire without prior neoantigen selection; however, efficacy can be weak when lower-affinity clones dominate the T cell pool. Chimeric Ag receptor T cell and tumor-infiltrating lymphocyte therapies additionally face challenges related to genetic modification, T cell exhaustion, and immunotoxicity. In this review, we highlight some engineering approaches and opportunities to these challenges among four classes of autologous cell therapies.
Collapse
Affiliation(s)
- Colleen R. Foley
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Sheridan L. Swan
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Melody A. Swartz
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
- Committee on Immunology, University of Chicago, Chicago, Illinois
- Ben May Department of Cancer Research, University of Chicago, Chicago, Illinois
| |
Collapse
|
2
|
Jiménez-Reinoso A, Nehme-Álvarez D, Domínguez-Alonso C, Álvarez-Vallina L. Synthetic TILs: Engineered Tumor-Infiltrating Lymphocytes With Improved Therapeutic Potential. Front Oncol 2021; 10:593848. [PMID: 33680923 PMCID: PMC7928359 DOI: 10.3389/fonc.2020.593848] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/30/2020] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy has emerged as an effective and life-changing approach for several types of cancers, both liquid and solid tumors. In combination with traditional treatments such as radiotherapy and/or chemotherapy, immune checkpoints inhibitors have improved prognosis and overall survival of patients with advanced melanoma and many other cancers. Among adoptive cell therapies (ACT), while chimeric antigen receptor T cell therapies have demonstrated remarkable efficacy in some hematologic malignancies, such as B cell leukemias, their success in solid tumors remains scarce due to the characteristics of the tumor microenvironment. On the other hand, ACT using tumor-infiltrating lymphocytes (TILs) is arguably the most effective treatment for metastatic melanoma patients, but even if their isolation has been achieved in epithelial tumors, their success beyond melanoma remains limited. Here, we review several aspects impacting TIL- and gene-modified “synthetic” TIL-based therapies and discuss future challenges that must be addressed with these approaches.
Collapse
Affiliation(s)
- Anaïs Jiménez-Reinoso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Daniel Nehme-Álvarez
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Carmen Domínguez-Alonso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Luis Álvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| |
Collapse
|
3
|
Strizova Z, Bartunkova J, Smrz D. The challenges of adoptive cell transfer in the treatment of human renal cell carcinoma. Cancer Immunol Immunother 2019; 68:1831-1838. [PMID: 31222485 PMCID: PMC11028041 DOI: 10.1007/s00262-019-02359-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/11/2019] [Indexed: 12/13/2022]
Abstract
Renal cell carcinoma (RCC) is one of the most lethal urologic malignancies. Its incidence continues to rise worldwide with a rate of 2% per year. Approximately, one-third of the RCC patients are diagnosed at advanced stages due to the asymptomatic nature of its early stages. This represents a great hurdle, since RCC is largely chemoresistant/radioresistant, and targeted therapy of mRCC still has limited efficacy. The 5-year survival rate of metastatic RCC (mRCC) is only around 10%. Adoptive cell transfer (ACT), a particular form of cell-based anticancer immunotherapy, is a promising approach in the treatment of mRCC. The vaccination principle, however, faces unique challenges that preclude the efficacy of ACT. In this article, we review the main challenges of ACT in the treatment of mRCC and describe multiple methods that can be used to overcome these challenges. In this respect, the ultimate purpose of this review is to provide a descriptive tool by which to improve the development of novel protocols for ACT of mRCC.
Collapse
Affiliation(s)
- Zuzana Strizova
- Institute of Immunology, 2nd Faculty of Medicine and Motol University Hospital, Charles University, V Uvalu 84, 150 06, Prague, Czech Republic
| | - Jirina Bartunkova
- Institute of Immunology, 2nd Faculty of Medicine and Motol University Hospital, Charles University, V Uvalu 84, 150 06, Prague, Czech Republic
| | - Daniel Smrz
- Institute of Immunology, 2nd Faculty of Medicine and Motol University Hospital, Charles University, V Uvalu 84, 150 06, Prague, Czech Republic.
| |
Collapse
|
4
|
Strizova Z, Taborska P, Stakheev D, Partlová S, Havlova K, Vesely S, Bartunkova J, Smrz D. NK and T cells with a cytotoxic/migratory phenotype accumulate in peritumoral tissue of patients with clear cell renal carcinoma. Urol Oncol 2019; 37:503-509. [PMID: 31030972 DOI: 10.1016/j.urolonc.2019.03.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 03/14/2019] [Accepted: 03/19/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Renal cell carcinoma (RCC) is the most lethal urologic malignancy with increasing incidence worldwide. The conventional treatment strategies for advanced or recurrent RCC are not efficient and show considerable toxicities. Adoptive cell transfer (ACT) has become a promising treatment option for multiple cancers, particularly in combination with other therapeutic approaches. ACT often utilizes extensively in vitro expanded tumor-infiltrating lymphocytes (TILs). However, TILs are a very heterogeneous mix of cell populations and only those populations that have a cytotoxic and migratory potential are thought to deliver a therapeutic impact in ACT. The identification and localization of these therapeutically potent populations are therefore needed. METHODS AND MATERIALS A total number of 57 tissue samples from 19 RCC patients who underwent radical nephrectomy was analyzed. The tissue samples were obtained from the tumor, peritumoral tissue, and the adjacent healthy renal tissue. The tissues were sliced, enzymatically dissociated into single cell suspensions and the obtained cells further analyzed by flow cytometry for the expression of markers of lymphocyte cytotoxicity - TRAIL and FasL, and a surrogate marker of lymphocyte migratory activity - PECAM-1. The analyzed data were next correlated with the clinical and histopathological data. RESULTS Non-clear cell RCC (non-ccRCC) tumors showed a significantly decreased tumor infiltration with TRAIL+FasL+ NK cells but elevated infiltration with FasL+PECAM-1+ T cells as compared with clear cell RCC (ccRCC) tumors. Further analyses revealed that the peritumoral tissue of ccRCC patients is a reservoir of TRAIL+FasL+, TRAIL+PECAM-1+, or FasL+PECAM-1+ NK and T cells. CONCLUSIONS The cytotoxic/migratory lymphocytes were identified in tumors of ccRCC patients. These lymphocytes became excluded from the tumor and accumulated in the patient's peritumoral tissue.
Collapse
Affiliation(s)
- Zuzana Strizova
- Institute of Immunology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Czech Republic
| | - Pavla Taborska
- Institute of Immunology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Czech Republic
| | - Dmitry Stakheev
- Institute of Immunology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Czech Republic
| | - Simona Partlová
- Institute of Immunology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Czech Republic
| | - Klara Havlova
- Department of Urology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Czech Republic
| | - Stepan Vesely
- Department of Urology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Czech Republic
| | - Jirina Bartunkova
- Institute of Immunology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Czech Republic
| | - Daniel Smrz
- Institute of Immunology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Czech Republic.
| |
Collapse
|
5
|
Abstract
Treatment and management of kidney disease currently presents an enormous global burden, and the application of nanotechnology principles to renal disease therapy, although still at an early stage, has profound transformative potential. The increasing translation of nanomedicines to the clinic, alongside research efforts in tissue regeneration and organ-on-a-chip investigations, are likely to provide novel solutions to treat kidney diseases. Our understanding of renal anatomy and of how the biological and physico-chemical properties of nanomedicines (the combination of a nanocarrier and a drug) influence their interactions with renal tissues has improved dramatically. Tailoring of nanomedicines in terms of kidney retention and binding to key membranes and cell populations associated with renal diseases is now possible and greatly enhances their localization, tolerability, and efficacy. This Review outlines nanomedicine characteristics central to improved targeting of renal cells and highlights the prospects, challenges, and opportunities of nanotechnology-mediated therapies for renal diseases.
Collapse
|
6
|
Groth A, Salnikov AV, Ottinger S, Gladkich J, Liu L, Kallifatidis G, Salnikova O, Ryschich E, Giese N, Giese T, Momburg F, Büchler MW, Moldenhauer G, Herr I. New Gene-Immunotherapy Combining TRAIL-Lymphocytes and EpCAMxCD3 Bispecific Antibody for Tumor Targeting. Clin Cancer Res 2012; 18:1028-38. [DOI: 10.1158/1078-0432.ccr-11-2767] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
7
|
de Bruyn M, Wei Y, Wiersma VR, Samplonius DF, Klip HG, van der Zee AGJ, Yang B, Helfrich W, Bremer E. Cell surface delivery of TRAIL strongly augments the tumoricidal activity of T cells. Clin Cancer Res 2011; 17:5626-37. [PMID: 21753155 DOI: 10.1158/1078-0432.ccr-11-0303] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Adoptive T-cell therapy generally fails to induce meaningful anticancer responses in patients with solid tumors. Here, we present a novel strategy designed to selectively enhance the tumoricidal activity of T cells by targeted delivery of TNF-related apoptosis-inducing ligand (TRAIL) to the T-cell surface. EXPERIMENTAL DESIGN We constructed two recombinant fusion proteins, anti-CD3:TRAIL and K12:TRAIL. Tumoricidal activity of T cells in the presence of these fusion proteins was assessed in solid tumor cell lines, primary patient-derived malignant cells, and in a murine xenograft model. RESULTS When added to T cells, K12:TRAIL and anti-CD3:TRAIL selectively bind to the T-cell surface antigens CD3 and CD7, respectively, leading to cell surface accretion of TRAIL. Subsequently, anti-CD3:TRAIL and K12:TRAIL increased the tumoricidal activity of T cells toward cancer cell lines and primary patient-derived malignant cells by more than 500-fold. Furthermore, T-cell surface delivery of TRAIL strongly inhibited tumor growth and increased survival time of xenografted mice more than 6-fold. CONCLUSIONS Targeted delivery of TRAIL to cell surface antigens of T cells potently enhances the tumoricidal activity of T cells. This approach may be generally applicable to enhance the efficacy of adoptive T-cell therapy.
Collapse
Affiliation(s)
- Marco de Bruyn
- Department of Surgery, Surgical Research Laboratories, University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Li YG, Wang ZP, Tian JQ, Tian BQ, Rodrigues R, Shang PF, Zhang T. Dendritic cell transfected with secondary lymphoid-tissue chemokine and/or interleukin-2 gene-enhanced cytotoxicity of T-lymphocyte in human bladder tumor cell S in vitro. Cancer Invest 2009; 27:909-17. [PMID: 19832038 DOI: 10.3109/07357900802375746] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES The aim of this study was to investigate whether dendritic cells (DCs) transfected with human secondary lymphoid-tissue chemokine (hSLC) and human interleukin-2 (hIL-2) genes are capable of improving DC's proliferation and to produce a marked antitumor effect in vitro combined with T-lymphocyte (TC). METHODS SLC gene primer was designed based on the corresponding gene sequence in GenBank. The Kpn I site was introduced into the upstream of the primer and Xho I site into the downstream. The SLC gene was amplified with the template of pET32a(+)-SLC by polymerase chain reaction. SLC was cloned into pBudCE4.1/IL-2 (TRAIL was cut from pBudCE4.1/TRAIL- IL-2 before) to construct recombinant plasmid pBudCE4.1/SLC-IL-2(PSI). DCs were transfected with pBudCE4.1/SLC-IL-2 by gene electric transfection. Protein expression was determined with Western blot and enzyme-linked immunosorbent assays. Cytotoxicity of TC and DC against the human bladder tumor cell were examined by chromium release assay. Flow cytometric analyses were performed to determine the apoptosis of tumor cells and the percentage of Treg. RESULTS A high level of expression of SLC and IL-2 was observed in DCs transfected with SLC and IL-2 genes. The mean production of IL-2 was 19.8 +/- 2.5, 511.10 +/- 52.36, and 541.3 +/- 62.04 ng/10(6) cells/24 hours in the DC/vector, DC/IL-2, and DC/SLC-IL-2, respectively. The mean SLC production was 29.8 +/- 4.43, 506.10 +/- 42.36, and 567.34 +/- 52.05 ngs/10(6)cells/24 hours in the DC/ vector, DC/SLC, and DC/SLC-IL-2, respectively. Cytotoxicity to bladder cancer cells was increased. The mean cytotoxicity (the effector/target ratio, 40:1) of TC-DC/parental, TC-DC/IL-2, TC-DC/SLC, and TC-DC/SLC-IL-2(TDSI) to the human bladder cancer cells was 32.1 +/- 5.5%, 63.5 +/- 6.6%, 78.1 +/- 9.63%, respectively. The apoptotsis rate of bladder cancer cells treated with TDSI was 18.6% by flow cytometry. Treg cells' percentage was very small in the DC medium. CONCLUSIONS SLC and IL-2 were produced by autocrine in DCs transfected with SLC and IL-2 genes. DC/SLC-IL-2 can promote DC proliferation, while TC-DC/SLC-IL-2 and TC-DC/SLC could strongly enhance significant cytotoxicity against bladder cancer cell that was induced by the coculture of DCs (transfected with SLC and IL-2) and TC.
Collapse
Affiliation(s)
- Yong-Gang Li
- Institute of Urology, Lanzhou University Second Hospital, Lanzhou University Clinical Medical College, Lanzhou, China
| | | | | | | | | | | | | |
Collapse
|
9
|
Macher-Goeppinger S, Aulmann S, Tagscherer KE, Wagener N, Haferkamp A, Penzel R, Brauckhoff A, Hohenfellner M, Sykora J, Walczak H, Teh BT, Autschbach F, Herpel E, Schirmacher P, Roth W. Prognostic value of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and TRAIL receptors in renal cell cancer. Clin Cancer Res 2009; 15:650-9. [PMID: 19147771 DOI: 10.1158/1078-0432.ccr-08-0284] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The death ligand tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its receptors (TRAIL-R) are involved in immune surveillance and tumor development. Here, we studied a possible association between the expression of TRAIL/TRAIL-Rs and the prognosis in patients with renal cell carcinomas (RCC). EXPERIMENTAL DESIGN A tissue microarray containing RCC tumor tissue samples and corresponding normal tissue samples from 838 patients was generated. Expression of TRAIL and TRAIL-Rs was examined by immunohistochemistry and the effect of TRAIL and TRAIL-R expression on disease-specific survival was assessed. RESULTS High TRAIL-R2 expression levels were associated with high-grade RCCs (P < 0.001) and correlated negatively with disease-specific survival (P = 0.01). Similarly, high TRAIL expression was associated with a shorter disease-specific survival (P = 0.01). In contrast, low TRAIL-R4 expression was associated with high-stage RCCs (P < 0.001) as well as with the incidence of distant metastasis (P = 0.03) and correlated negatively with disease-specific survival (P = 0.02). In patients without distant metastasis, multivariate Cox regression analyses revealed that TRAIL-R2 and TRAIL are independent prognostic factors for cancer-specific survival (in addition to tumor extent, regional lymph node metastasis, grade of malignancy, and type of surgery). CONCLUSION High TRAIL-R2, high TRAIL, and low TRAIL-R4 expression levels are associated with a worse disease-specific survival in patients with RCCs. Therefore, the assessment of TRAIL/TRAIL-R expression offers valuable prognostic information that could be used to select patients for adjuvant therapy studies. Moreover, our findings are of relevance for a potential experimental therapeutic administration of TRAIL-R agonists in patients with RCCs.
Collapse
|