Adoptive T Cell Transfer–Based Cancer Immunotherapy is a Promising Strategy for Cancer Treatment

Editorial

J Immun Res. 2014;1(1): 3.

Adoptive T Cell Transfer–Based Cancer Immunotherapy is a Promising Strategy for Cancer Treatment

Wang M* and Wang HY*

Center for Inflammation and Epigenetics, Houston Methodist Research Institute, USA

*Corresponding author: Wang M (or) Wang HY, Center for Inflammation and Epigenetics, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX77030, USA

Received: Aug 04, 2014; Accepted: Aug 05, 2014; Published: Aug 05, 2014

Editorial

Cancer is a leading cause of death worldwide [1]. According to the World Cancer Report 2014, the global burden of cancer rose to an estimated 14 million new cancer cases in 2012, and this figure is expected to rise to 22 million annually within the next two decades. Over the same period, cancer deaths are predicted to rise from an estimated 8.2 million to 13 million per year. Traditional cancer treatments, including surgery, chemotherapy, and radiation therapy, have demonstrated very limited efficacy for cancer patients with latestage disease.

The innate and acquired immune systems play a critical role in immune surveillance and immune defense [2,3]. Therefore, the use of the immune system to eliminate cancer is a very promising approach for cancer treatment [4, 5]. Indeed, immunotherapy has demonstrated great potential for cancer treatment [6–9], especially for disease refractory to traditional treatments. Recent FDA approval of immunotherapy–based vaccines/drugs sipuleucel–T (Provenge) [10] and ipilimumab (Yervoy) [11] represent milestones in the field of cancer immunotherapy for advanced prostate cancer and metastatic melanoma, respectively [8]. Furthermore, a phase III clinical trial of gp100 peptide vaccine in patients with advanced melanoma has shown encouraging results [12]. Thus, cancer immunotherapy has become an important part of treating cancer patients with advanced or refractory disease.

Cancer immunotherapy approaches include active immunization, reversal of immune suppression, nonspecific immune stimulation, and Adoptive Cell Transfer (ACT). To date, ACT has been demonstrated to be the most effective immunotherapy method for cancer treatment and has achieved very promising results in cancer clinical trials [13–17]. In the Tumor Infiltrating Lymphocyte (TIL)– based ACT approach, TILs are isolated from the tumor tissues of cancer patients, expanded in vitro using a high concentration of Interleukin (IL)–2 (6000 U/ml), and then infused back into the patient. The feasibility of the TIL–based ACT approach was first demonstrated in melanoma [18], with a current objective response rate of 49% to 72% when lymph depleting preparative regimen is performed prior to TIL infusion [7,19]. Despite the clinical benefits of TIL–based therapy, there are limitations to its successful implementation. TIL–based therapy is an individualized treatment that requires surgical removal of tumor tissues for TIL cultivation. Furthermore, few medical centers worldwide provide TIL–based therapy as it requires a highly skilled medical staff to isolate and cultivate TILs. To overcome these barriers, genetically modified cancer–specific T cells, such as T Cell Receptor (TCR)– and Chimeric Antigen Receptor (CAR)–transduced T cells are being developed to augment ACT–mediated immunotherapeutic responses against various types of cancer and have already shown encouraging therapeutic effects in clinical trials [13–17,20]. TCRs expressed on T cells can be genetically engineered in vitro to specifically recognize and kill cancer cells [15]. Cancer regression in patients with metastatic melanoma following administration of autologous T cells genetically engineered to express a TCR against MART1 was first reported in 2006 by Rosenberg et al. at the NIH [21]. TCR–transduced T cells against several tumor antigens including MART1, CEA, gp100, NY–ESO–1, and MAGEA3 have been tested in clinical trials [15]. The results from these clinical trials have shown great promise in treating various types of cancers including metastatic melanoma, metastatic colorectal cancer, metastatic synovial cell sarcoma, and epithelial malignancies. Importantly, CAR–transduced T cell based immunotherapy has been producing exciting clinical results. The concept of CAR was firstly introduced by Eshhar et al [22] and the CAR structure is composed of an extracellular single–chain variable fragment (scFv) of an antibody, a transmembrane domain, and intracellular signaling domains derived from molecules involved in T cell signaling. Although CAR–transduced T cells have been used to treat patients with various types of cancer in clinical trials, the most promising results from CAR–transduced T cell therapy have been with CD19–based targeting of B cell malignancies [23–27]. Several recent clinical trials performed by NIH, University of Pennsylvania, Memorial Sloan–Kettering Cancer Center have demonstrated positive clinical outcomes with anti–CD19 CAR T cell therapy in patients with B cell malignancies [6].

Although ACT–based cancer immunotherapy has exhibited encouraging results in clinical trials, this success has only been observed in a few types of cancer, predominantly in hematological cancers and melanoma. To develop safe and efficient ACT–based immunotherapy with broad efficacy against a wide range of cancer types, future research in ACT–based cancer immunotherapy should be focused on the following aspects: (1) to identify ideal cancer antigens. Currently, most clinical trials of ACT�based immunotherapy have used self–antigens that are over expressed in malignant cells compared with normal cells. Thus, infused antigenspecific T cells can target not only antigen–positive cancer cells in vivo but also normal tissues expressing the shared cancer antigen.The biggest challenge to overcome remains the identification of antigens with a strictly cancer cell–restricted expression, such as NYESO– 1, to achieve selective tumor targeting while sparing normal tissues. Mutated cancer antigens have been suggested as ideal targets for immune–based therapies, and such mutated cancer antigens can be identified by using a whole–exomic–sequencing–based approach [28,29]. (2) To enhance in vivo persistence and survival of adoptively transferred T cells. Results from clinical trials have suggested that cancer regression is positively associated with the long–term survival and persistence of adoptively transferred T cells in vivo. Strategies to enhance in vivo persistence and survival of transferred T cells can lead to improved anticancer efficacy. This may be achieved by exposure of T cells to IL–21 during in vitro culture [30], using Naïve CD8+ T cells or human stem cell–like memory T cells for adoptive immunotherapy [31,32], and by shRNA–mediated silencing of genes that inhibit T cell function in the tumor microenvironment[33]. Another alternative is to generate rejuvenated antigen–specific T cells that have a high proliferative capacity by reprogramming to pluripotency and redifferentiation [34,35]. (3) To identify cancer biomarkers for ACT–based therapy. Cancer biomarkers can support and guide clinical cancer treatment and development of the candidate products [36]. Thus, identification of biological correlates of response, either TILs or tumor characteristics, will help not only to select the most efficacious cell types for infusion but also patients who are most likely to respond to ACT–based therapy. (4) To use combinational immunotherapy. Although emerging techniques allow for the generation of high affinity antigen–specific T cells capable of targeting cancer cells in vitro, immune suppression and negative regulation at tumor sites can impede the induction of effective anticancer immune responses in vivo. Suppressive cytokines/agents such as IL– 10, transforming growth factor–β, and indoleamine 2,3–dioxygenase and cell populations such as MDSCs and Treg cells present in the tumor microenvironment induce immunosuppression, thereby blocking T cell function [37,38] and leading to poor antitumor efficacy of immunotherapy. Therefore, combining ACT therapy with other strategies that block negative regulators may induce potentially profound immune antitumor responses.

In conclusion, ACT–based cancer immunotherapy has achieved encouraging results in clinical trials and has curative potential in some cancer patients. These promising results obtained from clinicaltrials have earned cancer immunotherapy being named as the Science ‘Breakthrough of the Year’ in 2013 [39]. For the first time in many years, a number of pharmaceutical industries are investing heavily to facilitate the development of ACT–based cancer immunotherapy to treat various cancer types. We believe that the increasing interest and investments from pharmaceutical industries will accelerate research and development of ACT–based cancer immunotherapy to benefit patients with various types of cancers.

References

  1. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. See comment in PubMed Commons below CA Cancer J Clin. 2011; 61: 69–90.
  2. Vesely MD, Kershaw MH, Schreiber RD, Smyth MJ. Natural innate and adaptive immunity to cancer. See comment in PubMed Commons below Annu Rev Immunol. 2011; 29: 235–271.
  3. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. See comment in PubMed Commons below Science. 2011; 331: 1565–1570.
  4. Rosenberg SA. Cell transfer immunotherapy for metastatic solid cancer––what clinicians need to know. See comment in PubMed Commons below Nat Rev Clin Oncol. 2011; 8: 577–585.
  5. Wang HY, Wang RF. Enhancing cancer immunotherapy by intracellular delivery of cell–penetrating peptides and stimulation of pattern–recognition receptor signaling. See comment in PubMed Commons below Adv Immunol. 2012; 114: 151–176.
  6. Kochenderfer JN, Rosenberg SA. Treating B–cell cancer with T cells expressing anti–CD19 chimeric antigen receptors. See comment in PubMed Commons below Nat Rev Clin Oncol. 2013; 10: 267–276.
  7. Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: harnessing the T cell response. Nat Rev Immunol. 2012; 12: 269–281.
  8. Sharma P, Wagner K, Wolchok JD, Allison JP. Novel cancer immunotherapy agents with survival benefit: recent successes and next steps. Nat Rev Cancer. 2011; 11: 805–812.
  9. Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature. 2011; 480: 480–489.
  10. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. Sipuleucel–T immunotherapy for castration–resistant prostate cancer. N Engl J Med. 2010; 363: 411–422.
  11. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010; 363: 711–723.
  12. Schwartzentruber DJ, Lawson DH, Richards JM, Conry RM, Miller DM, Treisman J, et al. gp100 peptide vaccine and interleukin–2 in patients with advanced melanoma. N Engl J Med. 2011; 364: 2119–2127.
  13. Ruella M, Kalos M. Adoptive immunotherapy for cancer. Immunol Rev. 2014; 257: 14–38.
  14. Jensen MC, Riddell SR. Design and implementation of adoptive therapy with chimeric antigen receptor–modified T cells. Immunol Rev. 2014; 257: 127–144.
  15. Hinrichs CS, Rosenberg SA. Exploiting the curative potential of adoptive T–cell therapy for cancer. Immunol Rev. 2014; 257: 56–71.
  16. Dotti G, Gottschalk S, Savoldo B, Brenner MK. Design and development of therapies using chimeric antigen receptor–expressing T cells. Immunol Rev. 2014; 257: 107–126.
  17. Cheadle EJ, Gornall H, Baldan V, Hanson V, Hawkins RE, Gilham DE. CAR T cells: driving the road from the laboratory to the clinic. Immunol Rev. 2014; 257: 91–106.
  18. Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, et al. Use of tumor–infiltrating lymphocytes and interleukin–2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med. 1988; 319: 1676–1680.
  19. Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T–cell transfer immunotherapy. Clin Cancer Res. 2011; 17: 4550–4557.
  20. Yee C. The use of endogenous T cells for adoptive transfer. Immunol Rev. 2014; 257: 250–263.
  21. Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science. 2006; 314: 126–129.
  22. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin–T–cell receptor chimeric molecules as functional receptors with antibody–type specificity. Proc Natl Acad Sci USA. 1989; 86: 10024–10028.
  23. Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, et al. CD19–Targeted T Cells Rapidly Induce Molecular Remissions in Adults with Chemotherapy–Refractory Acute Lymphoblastic Leukemia. Sci Transl Med. 2013; 5.
  24. Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014; 6.
  25. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, et al. Chimeric antigen receptor–modified T cells for acute lymphoid leukemia. N Engl J Med. 2013; 368: 1509–1518.
  26. Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, et al. B–cell depletion and remissions of malignancy along with cytokine–associated toxicity in a clinical trial of anti–CD19 chimeric–antigen–receptor–transduced T cells. Blood. 2012; 119: 2709–2720.
  27. Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Stetler–Stevenson M, Feldman SA, et al. Eradication of B–lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood. 2010; 116: 4099–4102.
  28. Robbins PF, Lu YC, El–Gamil M, Li YF, Gross C, Gartner J, et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor–reactive T cells. Nat Med. 2013; 19: 747–752.
  29. Tran E, Turcotte S, Gros A, Robbins PF, Lu YC, Dudley ME, et al. Cancer immunotherapy based on mutation–specific CD4+ T cells in a patient with epithelial cancer. Science. 2014; 344: 641–645.
  30. Yang S, Ji Y, Gattinoni L, Zhang L, Yu Z, Restifo NP, et al. Modulating the differentiation status of ex vivo–cultured anti–tumor T cells using cytokine cocktails. Cancer Immunol Immunother. 2013; 62: 727–736.
  31. Gattinoni L, Lugli E, Ji Y, Pos Z, Paulos CM, Quigley MF, et al. A human memory T cell subset with stem cell–like properties. Nat Med. 2011; 17: 1290–1297.
  32. Hinrichs CS, Borman ZA, Gattinoni L, Yu Z, Burns WR, Huang J, et al. Human effector CD8+ T cells derived from naive rather than memory subsets possess superior traits for adoptive immunotherapy. Blood. 2011; 117: 808–814.
  33. Zhou P, Shaffer DR, Alvarez Arias DA, Nakazaki Y, Pos W, Torres AJ, et al. In vivo discovery of immunotherapy targets in the tumour microenvironment. Nature. 2014; 506: 52–57.
  34. Vizcardo R, Masuda K, Yamada D, Ikawa T, Shimizu K, Fujii S, et al. Regeneration of human tumor antigen–specific T cells from iPSCs derived from mature CD8(+) T cells. Cell Stem Cell. 2013; 12: 31–36.
  35. Nishimura T, Kaneko S, Kawana–Tachikawa A, Tajima Y, Goto H, Zhu D,et al. Generation of rejuvenated antigen–specific T cells by reprogramming to pluripotency and redifferentiation. Cell Stem Cell. 2013; 12: 114–126.
  36. Kalos M. Biomarkers in T cell therapy clinical trials. J Transl Med. 2011; 9: 138.
  37. Wang HY, Wang RF. Regulatory T cells and cancer. Curr Opin Immunol. 2007; 19: 217–223.
  38. Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol. 2010; 10: 490–500.
  39. Couzin–Frankel J. Breakthrough of the year 2013. Cancer immunotherapy. Science. 2013; 342: 1432–1433.

Download PDF

Citation: Wang M and Wang HY. Adoptive T cell Transfer–Based Cancer Immunotherapy is a Promising Strategy for Cancer Treatment. J Immun Res. 2014;1(1): 3. ISSN:2471-0261

Home
Journal Scope
Online First
Current Issue
Editorial Board
Instruction for Authors
Submit Your Article
Contact Us