Adoptive Cellular Therapy With Synthetic T Cells as an “Instant Vaccine” for Cancer and Immunity

Adoptive Cellular Therapy With Synthetic T Cells as an “Instant Vaccine” for Cancer and Immunity

Chapter 30 Adoptive Cellular Therapy With Synthetic T Cells as an “Instant Vaccine” for Cancer and Immunity Carl H. June, MD Center for Cellular Immu...

706KB Sizes 0 Downloads 95 Views

Chapter 30

Adoptive Cellular Therapy With Synthetic T Cells as an “Instant Vaccine” for Cancer and Immunity Carl H. June, MD Center for Cellular Immunotherapies, Abramson Cancer Center; University of Pennsylvania, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Philadelphia, PA, United States

Chapter Outline 1 Introduction 1.1 History of Adoptive Cell Transfer 1.2 Lymphocyte Cell Transfer 1.3 Emergence of ImmunoOncology 2 Discovery of T Cells with Stem-Cell-Like Qualities 3 Approaches to Engineer Lymphocytes Using Synthetic Biology

581 582 582 583 584

584

4 Design of CAR T Cells 5 Lessons from Clinical trials on Patients with Chronic Infection 6 Clinical trials with CAR T Cells Directed Against B-Cell Malignancies 7 Toxicities and Management 8 Conclusions and Future Challenges References

586

587

588 589 590 590

1 INTRODUCTION The fields of vaccinology and adoptive cell transfer (ACT) converge in the shaping of the acquired cellular and humoral responses of the B- and T cells of the immune system toward desired antigens on pathogens or self-targets. However, the approaches differ markedly, with vaccines often requiring immunizations and boosting over a period of months, whereas ex vivo culture of T cells and gene transfer can generate sufficient T cells in less than a week (Fig. 30.1). The process of ACT is more complex but the power of the resultant T cells may justify the expense and effort in some areas such as cancer and chronic infection, where vaccines often fail. The Vaccine Book. http://dx.doi.org/10.1016/B978-0-12-802174-3.00030-8 Copyright © 2016 Elsevier Inc. All rights reserved.

581

582

PART | XI  Future Challenges

FIGURE 30.1  General approach for trials with ACT. The patient donates lymphocytes by phlebotomy, and the cells are genetically modified and expanded in numbers in the manufacturing facility. In an optional step, the patient may receive chemotherapy before intravenous infusion of the CAR T cells.

1.1  History of Adoptive Cell Transfer A scientific and technological revolution in the war against cancer has taken place in the past century. Paul Ehrlich was probably the first to envision targeted therapies with “magic bullets,”1 an idea that was actualized by the invention of monoclonal antibodies and has inspired generations of scientists to devise powerful molecular cancer therapeutics. In this review the potential uses of chimeric T cells, modified to express antibody fragments is discussed. The idea that passive transfer of primed immune cells can generate immunity in the recipient of this transfer is a relatively old idea in the history of immunology. As first proposed by Billingham, Brent, and Medawar in 1954, who coined the term “adoptive immunity,”2 numerous animal studies have demonstrated the effectiveness of adoptive transfer of immunity toward cancer and infectious disease. Immunity has remarkable specificity toward its targets, and specificity can be controlled through strategies such as in vivo and ex vivo priming and genetic engineering to install receptors of defined specificities. Moreover, it has the potential to induce longstanding effects via the establishment of immunologic memory.

1.2  Lymphocyte Cell Transfer In early studies, the transfer of pig and nonhuman primate lymphocytes were tested and found inadequate for ACT due to transient engraftment.3,4 However,

Adoptive Cellular Therapy Chapter | 30

583

with the advent of efficient tools for genetic editing, it is not inconceivable that the use of xenogeneic cells for ACT might be reconsidered. Currently, human T cells with alpha beta T-cell receptors (TCRs) are currently the cell of choice for most clinical trials, reviewed in Ref. 5. Gene transfer of MHC class I-restricted TCRs can “convert” a population of polyclonal CD8+ T cells to cytotoxic Tlymphocytes (CTLs) of monoclonal TCR specificity.6 The adoptive transfer of engineered CD4+ T cells has promise for adoptive therapy of cancer and HIV, reviewed in Refs. 7,8. Recent advances in the understanding of the biology of γδ T cells suggest that these cells have promise for ACT. For example, human γδ T cells can prime αβ T cells with an efficiency similar to that of dendritic cells.9 Human γδ T cells can be engineered to express chimeric antigen receptors (CARs) and αβ TCRs.10,11 The use of γδ T cells may provide a significant safety feature over the use of TCR-engineered αβ T cells, taking advantage of the finding that α and β TCR chains cannot pair with γ and δ TCR chains. Natural killer T (NKT) cells are functionally related to γδ T cells since they also bridge innate and adaptive immune responses and can enhance or suppress immunity.12 The best characterized human NKT cell subpopulation, referred to as invariant NKT (iNKT) cells, expresses CD161 and an invariant Vα24Jα18 TCR that recognizes α-galactosylceramide presented by the MHC class I-like molecule CD1d. After activation, iNKT cells have MHC-independent cytotoxic activity against various tumors and secrete high levels of interferon-γ, although this function becomes impaired in patients with cancer.13 Compared with mouse NKT cells, human NKT cells are rare and comprise <1% of total lymphocytes. However, human NKT cells, unlike mouse NKT cells, can undergo substantial expansion in vitro.14 The first ACT studies of iNKT cells have been conducted and show safety and some evidence of antitumor activity.15

1.3  Emergence of Immuno-Oncology Targeting of disease through the adoptive transfer of lymphocytes was first reported more than 50 years ago in rodent models.16 However, it is only recently that the widespread use of immune-oncology has been accepted into the daily practice of medicine. This is best illustrated by the striking success of immune checkpoint blocking antibodies, which have achieved the complete and sustained remission of advanced solid tumors,17 and the use of genetically engineered T cells for melanoma and leukemia.18 The field of ACT has been made possible by improved understanding of T-cell biology, including the mechanisms for T-cell activation and recognition of targets, the role of accessory surface molecules and signal transduction pathways involved in the regulation of T-cell function and survival, as well as the identification and cloning of soluble T-cell growth factors, has facilitated the ability to expand ex vivo large numbers of highly potent T cells for adoptive immunotherapy. There are several excellent reviews of the experimental basis for ACT therapy of tumors and chronic infections.19–21

584

PART | XI  Future Challenges

2  DISCOVERY OF T CELLS WITH STEM-CELL-LIKE QUALITIES Mouse syngeneic tumor models have been essential for the identification and preclinical optimization of many tumor therapies. However, mouse tumor models in general have not been a good predictor of responses to ACT for cancer although they have been quite useful for chronic infections. In part this is likely due to the fact that most tumor models in mice don’t mirror the mutational loads and chronic inflammation found in humans, and due to the fact that the onset human T-cell senescence is quite different from mice, reviewed in Ref. 20. The seminal discovery of telomerase22 and the developing field of telomere biology identified fundamental differences in mechanisms of immune senescence between mice and humans23–25 that have important implications for ACT. In human T cells, a decrease in mean telomere restriction fragment length was shown for increasing age.26,27 As one consequence of the appreciation of the differences in biology between mouse and human T cells, the duration of culture for human T cells used in adoptive transfer has decreased from a month or longer to days to a few weeks, resulting in preservation of telomere length in infused T cells. More recent studies in the mouse indicate that a memory stem cell subset of CD8+ T cells exists.28 Similar T cells with stem cell-like qualities have been identified in humans.29,30 In elegant and technically demanding studies, these memory stem T cells were shown to have plasticity and that a single naive T cell can reconstitute diverse effector and memory T-cell subsets in mice.31,32 Ongoing studies in human allotransplant patients indicate that infusions of very low numbers of CMV-specific T cells are able to provide protective immunity.33 Efficient systems for the growth of human T cells were developed based on principles of costimulation.34,35 When these culture systems were employed in patients with HIV infection, efficient engraftment and long-term persistence of T cells was demonstrated after ACT.36,37 CD4+ T cells rendered CCR5-deficient by zinc finger nuclease (ZFN) editing also have been shown to persist for at least 5 years after ACT in patients with chronic HIV infection, consistent with the existence of central memory T cells in humans with long life spans.38

3  APPROACHES TO ENGINEER LYMPHOCYTES USING SYNTHETIC BIOLOGY In contrast to promising results in patients with chronic infection, ACT with natural T cells was not generally shown to be beneficial in cancer patients. A major reason for this is that tolerance to tumor antigens is induced by a variety of mechanisms, including deletion of tumor reactive cells and the induction of adaptive resistance mediated by checkpoint molecules such as CTLA-4 and PD1.39

Adoptive Cellular Therapy Chapter | 30

585

Synthetic biology, an emerging discipline aimed at reprogramming living organisms through the combined use of genetics, engineering principles, and systems and computational analysis, is able to enhance T-cell behavior.40–42 Cells are inherently capable of carrying out complex computations and responses, and CTLs of the immune system in particular are composed of cells poised to kill tumor cells after through careful assessment of targets. Approaches to genetically engineered lymphocytes have been reviewed previously.5 Using the principles of gene transfer it is possible to change “at will” the specificity of T cells using CAR and TCR of known specificity and affinity. Additional strategies have involved the overexpression in T cells of prosurvival signals such as telomerase,43 antiapoptotic genes,44 and the downregulation of proapoptotic molecules such as Fas.45 Yet another approach to enhance T-cell survival involves the expression of dominant-negative receptors for inhibitory molecules such as dominant-negative receptors for TGF-beta.46 Over the past decade a number of tools including ZFN, transcription activator-like effector nucleases (TALENs) and clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 system (CRISPER/ Cas9) have been developed to permit gene-specific disruption and site-specific insertion of novel DNA sequences.47 To date only ZFNs have been used for human trials.38 However, in vitro these tools are enabling the discovery of genes that increase the function by T cells by loss of function or gain of function mutations. For example, Zhou and coworkers48 reported a novel RNA-interference based screen conducted in vivo using ACT with transduced TCR transgenic T cells, with enrichment of shRNAs occurring in T cells whose numbers in the tumor bed increased upon knock down of relevant genes. Others used a CRISPR library approach for the in vitro analysis of LPS signaling in dendritic cells.49 Both screens identified known as well as unknown targets that affected the response being assessed. Using CRISPR/Cas9 technology, Konermann and coworkers reported a discovery method testing selective genome-wide gene activation to achieve desired phenotypes.50 Together, these new tools are overcoming a key challenge facing the cancer immunology field in the discovery of the most suitable targets to synthetically “hack” T cell to enhance T-cell antitumor functions. The diverse approaches to effectively engineer T cells combined with recently acquired mechanistic insights into T-cell biology and tumor immunity have converged to the point where the rational engineering of potent antitumor T-cell immunity is a practical and clinically testable reality. The majority of clinical approaches have employed T cells engineered to stably express transgenes via virus-based transduction. Virus-mediated gene transfer approaches typically employ vectors that are derived from gamma retroviruses or more recently lentiviruses, principally because of their ability to integrate into the host genome and drive long-term transgene expression, as well as for their low intrinsic immunogenicity. Gammaretrovirus-based transduction requires replicating cells for viral integration into genomic DNA, whereas lentiviral vectors

586

PART | XI  Future Challenges

can also integrate into nondividing cells; lentiviral vectors also appear to be less susceptible to silencing by host restriction factors and can deliver larger DNA sequences than retroviruses.51 Although virus-based approaches result in reasonably efficient transduction of primary T cells, they have considerable limitations in terms of cost to manufacture clinical-grade material, the total size of DNA that can be included in the virus vectors, and the potential, principally for retroviruses, for the integration events to result in insertional oncogenesis. A new virus-based system that has not yet entered clinical trials is based on foamy virus vectors, which possess favorable integration properties and are not pathogenic in humans.52 Nonvirus-based approaches benefit from lower manufacturing cost and are in principle less immunogenic than viral approaches. Although such approaches are theoretically safer because they are not dependent on viral elements integrating into host DNA, their safety record is shorter than that of virus-based vectors. Nonviral approaches to introduce transgenes into T cells involve the utilization of transposon elements such as sleeping beauty and piggybac as well as ZFN, TALEN, and CRISPR/Cas9 based-technologies, which allow for the ability to engineer T-cell populations with transgene insertions into specific chromosomal loci or that are biallelically disrupted for specific genes.53–57 Such technologies offer significant potential to engineer T cells in a manner that allows for the ability to interrupt or otherwise modulate expression of particular proteins that may be deleterious to therapeutic function.

4  DESIGN OF CAR T CELLS CARs are synthetic, engineered receptors that can target surface molecules in their native conformation.58,59 Unlike TCRs, CARs engage molecular structures independent of antigen processing by the target cell and independent of MHC; recognition features that are desirable as an approach to overcome resistance mechanisms found in tumors and chronic infection. CARs typically engage the target via a single-chain variable fragment (scFv) derived from an antibody, although natural ligands have also been used.60 Individual scFv’s targeting a surface molecule are derived either from murine or humanized antibodies, or synthesized and identified by screening of phage display libraries.61 Unlike TCRs, where a narrow range of affinity dictates the activation and specificity of the T cell, CARs typically have a much higher and perhaps broader range of affinities that will engage the target without necessarily encountering crossreactivity issues. Varying the affinity can increase the therapeutic index of CAR T cells, by increasing the discrimination of normal tissue expression from levels of surrogate antigen expressed on tumors.62,63 The length, flexibility, and origin of the hinge domain is also an important variable in the design of CARs.62,64,65 A major challenge to the field is that it is currently necessary to empirically test these design variables as there are no general rules guiding CAR design for selected target molecules.

Adoptive Cellular Therapy Chapter | 30

587

FIGURE 30.2  CAR design. In the natural immune system, B cells display antibodies on their surface and T cells have TCRs on the surface. A synthetic CAR T cell is a chimera of a B- and T cell, displaying both an antibody fragment (scFv) and an endogenous TCR.

The “generations” of CARs typically refer to the number and composition intracellular signaling domains (Fig. 30.2). “First-generation” CARs include only CD3zeta as an intracellular signaling domain whereas second-generation CARs also include costimulatory domains. Most investigators are currently using the hinge and transmembrane domains of CD8alpha or CD28. Hinge domains derived from Fc regions have also been investigated and modified in length,62 and in some instances these hinges have also been reported to engage Fc receptors and activate innate immune cells.65

5  LESSONS FROM CLINICAL TRIALS ON PATIENTS WITH CHRONIC INFECTION Adoptive immunotherapy began in humans shortly after the discovery and production of IL-2 (Table 30.1). The initial trials in cancer patients were largely unsuccessful66; the reasons for the failure of the initial trials have only recently become apparent and include quantitative and qualitative deficiencies in the repertoire of the infused TCRs, infusion of cells that had reached replicative senescence, and susceptibility to checkpoint pathways in vivo that prevented responses. However, in patients with chronic viral infections, ACT was shown to have promise in early trials as ACT with natural CTL clones specific for cytomegalovirus and Epstein–Barr virus infection were shown to be effective in immunosuppressed individuals.67,68 Substantial data exist to indicate that CD8+ T cells can affect the outcome and viral load in HIV-1 infection. Naturally occurring gag-specific CTL responses are inversely associated with viremia.69 In contrast, adoptive CTL therapy for HIV/AIDS, though demonstrating safety and promising engraftment and trafficking of cells to sites of viral replication, has not been clinically effective,70 and is accompanied by rapid emergence of viral escape mutants.71 Mathematical modeling suggests that adoptive transfer of CTLs should augment HIV-1 immunity and control viral replication, but only

588

PART | XI  Future Challenges

TABLE 30.1 Potential Indications for Various Forms of ACT Therapies Indication

Cell type

References

Metastatic melanoma

Tumor infiltrating lymphocytes

[76]

B-cell lineage tumors

CD19 CAR T cells

[77]

Synovial cell sarcoma Multiple myeloma

NY-ESO-1 TCR T cells

[78] [79]

Lymphopenia/immune reconstitution

Polyclonal T cells

[36]

Donor lymphocyte infusions

Allogeneic T cells:

[80]

Type 1 diabetes

Autologous Tregs

[81]

Prevention of organ transplant rejection

Inducible and natural Tregs

[82]

Graft versus host disease

Cord blood derived donor Tregs

[83]

HIV: increase immunity

CTL clones

[84]

HIV: increase immunity

CAR T

[85]

HIV: increase resistance

CCR5-deficient T cells

[38]

CMV: therapy for drug-resistant virus

CTLs

[33,84]

EBV: therapy for drug-resistant virus

CTLs

[68]

when the replicative capacity of the genetically modified CTLs is preserved and functional CD4+ T cells are present.72,73 Thus, an attractive strategy is the use of genetically enhanced TCRs to facilitate immune-mediated control of viral replication.74 Ultimately, a two-pronged approach involving gene-modified T cells comprising CD4+ T-cell protection and CTL augmentation therapy might be optimal.75

6  CLINICAL TRIALS WITH CAR T CELLS DIRECTED AGAINST B-CELL MALIGNANCIES Maus and coworkers reviewed the literature and listed all CAR T-cell trials up to late in 2013.86 A search of the clinicaltrials.gov data base reveals that there are 88 trials testing CAR T cells that are currently registered.a Geographically most of the trials are being conducted in the United States, although China is now the second most prevalent site of investigation with CAR T cells. Although the earliest trials of CAR T-cell therapy were performed in patients with solid tumors,87,88 these trials were disappointing. It is actually in trials of patients with B-cell malignancies that the most exciting results have recently been obtained.77,89–92 a. Search terms “chimeric antigen receptor,” Dec. 10, 2015.

Adoptive Cellular Therapy Chapter | 30

589

Many groups including those at Memorial Sloan Kettering, Seattle Children’s and the Fred Hutchinson Cancer Research Center, the National Cancer Institute, and others, have reported clinical responses in relapsed refractory leukemia and lymphoma. At our center, we have treated more than 200 patients with CAR T cells targeting B-cell malignancies including chronic lymphoid leukemia, acute lymphoid leukemia, non-Hodgkin’s lymphoma, and myeloma. B-cell malignancies are particularly amenable to be targeted using CAR T-cell therapy, due to the presence of the CD19 and CD20 antigens on most B-cell malignancies from the most immature B-cell acute lymphoblastic leukemia (B-ALL) to the most mature lymphomas93 and the fact that patients can tolerate prolonged periods of B-cell aplasia. Treatment with CAR T cells specific for CD19 has resulted in complete responses in B-ALL and chronic lymphocytic leukemia. However, one recurrent observation among trials in B-ALL is the emergence of CD19-negative blasts at relapse in a substantial minority (approximately 15%) of patients.91,92,94 Loss of CD19 is rarely observed in CLL, suggesting that CD19 escape is more difficult, or that CD19 negative precursor cells for CLL do not occur. Fewer published studies are currently available for comparison of response rates against mature B-cell neoplasms and non-Hodgkin lymphoma and no examples of relapse attributable to CD19-negative escape variants have yet been reported. Myeloma has been successfully targeted in a pilot trial with CD19 CAR T cells,95 an event that was unexpected since myeloma typically does not express CD19. After treatment with B cell–directed CAR T cells, the response rates vary by disease. In CLL we observe an approximate 60% overall response rate in relapsed/refractory patients.96 In contrast, in young adults and children with ALL who have already failed a stem-cell transplant, there is a remarkable 90% complete response rate.94,97 There are some late relapses, largely attributed to either loss of functional CAR T cells in the patient, or to an emergence of a leukemia clone that does not express the CD19 target antigen that is recognized by the CAR T cell.98 The emerging solution to this is to have another “CAR in the garage” targeting other markers on B cells, a strategy that we have just begun testing with CD22-specific CAR T cells.

7  TOXICITIES AND MANAGEMENT In most cases the toxicities associated with CAR T cells are on-target and reversible, a characteristic setting this treatment apart from nearly all previous cancer therapies comprised of cytotoxic chemotherapy and radiation therapy.99 These toxicities include B-cell aplasia, tumor lysis syndrome, cytokine release syndrome, and macrophage activation syndrome.89,100 Hyperferritinemia and elevations in serum C-reactive protein are observed in patients with cytokine release syndrome and macrophage activation syndrome.91,97 The occurrence of B-cell aplasia and duration of persistence of CAR T cells in patients with leukemia is a predictive biomarker of response to therapy.94,96 Cytokine release

590

PART | XI  Future Challenges

syndrome has been reported by all groups after treatment of B-cell malignancies, and has also been observed after the treatment of multiple myeloma,95 which is typically considered a CD19 negative malignancy. Cytokine release syndrome is now effectively managed with tocilizumab and other reagents that block IL-6 signaling.91,101 The use of cytokine blockage may be preferable to immunosuppression with corticosteroids due to enhanced therapeutic effects and less systemic immunosuppression.102 CNS symptoms and signs consisting of expressive aphasia, confusion, and seizures have been observed after CD19 CAR therapy.103 This syndrome is reversible and the cause of these syndrome remains unexplained because there is no obvious correlation with symptoms and tumor in the CNS. This may be a drug class effect because the syndrome is also observed after treatment with blinatumomab, a CD19 directed bispecific single-chain antibody.104

8  CONCLUSIONS AND FUTURE CHALLENGES Adoptive immunotherapy using autologous T cells endowed with CARs has emerged as a powerful approach to treating cancer. However, a current limitation of this approach is that autologous CAR T cells must be generated on a bespoke basis. The development of automated culture technologies employing robotics will be required to scale out this therapy for general incorporation into the routine practice of medicine.105 Further, the potential development of third party cells through large-scale manufacturing of T cells deficient in expression of their TCR and other molecules will likely ameliorate this issue through the use of “off-the-shelf” CAR T cells.106 There are now dozens of cell therapy companies and it is likely that this emerging industry will solve issues of manufacturing feasibility.18 The major scientific issue in the field is whether engineered T cells will have substantial impact in adenocarcinoma and other solid tumors; at this point efficacy following adoptive transfer is limited and most routinely observed with natural tumor infiltrating lymphocytes and TCR T cells rather than CAR T cells.107 Finally, the ability to mass produce genetically modified T cells with desired specificities has the potential to enable “instant vaccines” as a form of immunotherapy that can be effective rapidly and in immunosuppressed patients, where standard vaccines are often ineffective.

REFERENCES 1. Strebhardt K, Ullrich A. Paul Ehrlich’s magic bullet concept: 100 years of progress. Nat Rev Cancer 2008;8(6):473–80. 2. Billingham RE, Brent L, Medawar PB. Quantitative studies on tissue transplantation immunity. II. The origin, strength and duration of actively and adoptively acquired immunity. Proc Royal Soc 1954;143:58–80. 3. Symes MO, Riddell AG. The use of immunized pig lymph-node cells in the treatment of patients with advanced malignant disease. Br J Surg 1973;60(3):176–80.

Adoptive Cellular Therapy Chapter | 30

591

4. Michaels MG, Kaufman C, Volberding PA, Gupta P, Switzer WM, Heneine W, et al. Baboon bone-marrow xenotransplant in a patient with advanced HIV disease: case report and 8-year follow-up. Transplantation 2004;78(11):1582–9. 5. June CH, Blazar BR, Riley JL. Engineering lymphocyte subsets: tools, trials and tribulations. Nat Rev Immunol 2009;9(10):704–16. 6. Cooper LJ, Kalos M, Lewinsohn DA, Riddell SR, Greenberg PD. Transfer of specificity for human immunodeficiency virus type 1 into primary human T lymphocytes by introduction of T-cell receptor genes. J Virol 2000;74(17):8207–12. 7. Rossi JJ, June CH, Kohn DB. Genetic therapies for HIV/AIDS. Nat Biotechnol 2007;25(12):1444–54. 8. Muranski P, Restifo NP. Adoptive immunotherapy of cancer using CD4(+) T cells. Curr Opin Immunol 2009;21(2):200–8. 9. Brandes M, Willimann K, Moser B. Professional antigen-presentation function by human gammadelta T cells. Science 2005;309(5732):264–8. 10. Rischer M, Pscherer S, Duwe S, Vormoor J, Jurgens H, Rossig C. Human gammadelta T cells as mediators of chimaeric-receptor redirected anti-tumour immunity. Br J Haematol 2004;126(4):583–92. 11. van der Veeken LT, Coccoris M, Swart E, Falkenburg JH, Schumacher TN, Heemskerk MH. Alpha beta T cell receptor transfer to gamma delta T cells generates functional effector cells without mixed TCR dimers in vivo. J Immunol 2009;182(1):164–70. 12. Kronenberg M. Toward an understanding of NKT cell biology: progress and paradoxes. Annu Rev Immunol 2004;23:877–900. 13. Tahir SM, Cheng O, Shaulov A, Koezuka Y, Bubley GJ, Wilson SB, et al. Loss of IFN-gamma production by invariant NK T cells in advanced cancer. J Immunol 2001;167(7):4046–50. 14. Exley MA, Hou R, Shaulov A, Tonti E, Dellabona P, Casorati G, et al. Selective activation, expansion, and monitoring of human iNKT cells with a monoclonal antibody specific for the TCR a-chain CDR3 loop. Eur J Immunol 2008;38(6):1756–66. 15. Motohashi S, Nagato K, Kunii N, Yamamoto H, Yamasaki K, Okita K, et al. A phase I-II study of alpha-galactosylceramide-pulsed IL-2/GM-CSF-cultured peripheral blood mononuclear cells in patients with advanced and recurrent non-small cell lung cancer. J Immunol 2009;182(4):2492–501. 16. Mitchison NA. Studies on the immunological response to foreign tumor transplants in the mouse. I. The role of lymph node cells in conferring immunity by adoptive transfer. J Exp Med 1955;102:157–77. 17. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366(26):2443–54. 18. June CH, Riddell SR, Schumacher TN. Adoptive cellular therapy: a race to the finish line. Sci Transl Med 2015;7(280):280ps7. 19. Riddell SR, Greenberg PD. Principles for adoptive T cell therapy of human viral diseases. Annu Rev Immunol 1995;13:545–86. 20. June CH. Principles of adoptive T cell cancer therapy. J Clin Invest 2007;117(5):1204–12. 21. June CH. Adoptive T cell therapy for cancer in the clinic. J Clin Invest 2007;117(6):1466–76. 22. Blackburn EH, Greider CW, Szostak JW. Telomeres and telomerase: the path from maize, Tetrahymena and yeast to human cancer and aging. Nat Med 2006;12(10):1133–8. 23. Forsyth NR, Wright WE, Shay JW. Telomerase and differentiation in multicellular organisms: turn it off, turn it on, and turn it off again. Differentiation 2002;69(4–5):188–97. 24. Akagi T. Oncogenic transformation of human cells: shortcomings of rodent model systems. Trends Mol Med 2004;10(11):542–8.

592

PART | XI  Future Challenges

25. Horikawa I, Chiang YJ, Patterson T, Feigenbaum L, Leem SH, Michishita E, et al. Differential cis-regulation of human versus mouse TERT gene expression in vivo: identification of a human-specific repressive element. Proc Natl Acad Sci USA 2005;102(51):18437–42. 26. Weng NP, Palmer LD, Levine BL, Lane HC, June CH, Hodes RJ. Tales of tails: regulation of telomere length and telomerase activity during lymphocyte development, differentiation, activation, and aging. Immunol Rev 1997;160:43–54. 27. Weng NP, Levine BL, June CH, Hodes RJ. Human naive and memory T lymphocytes differ in telomeric length and replicative potential. Proc Natl Acad Sci USA 1995;92:11091–4. 28. Zhang Y, Joe G, Hexner E, Zhu J, Emerson SG. Host-reactive CD8+ memory stem cells in graft-versus-host disease. Nat Med 2005;11(12):1299–305. 29. Turtle C, Swanson H, Fujii N, Estey E, Riddell S. A distinct subset of self-renewing human memory CD8+ T cells survives cytotoxic chemotherapy. Immunity 2009;31(5):834–44. 30. 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(10):1290–7. 31. Buchholz VR, Flossdorf M, Hensel I, Kretschmer L, Weissbrich B, Graf P, et al. Disparate individual fates compose robust CD8+ T cell immunity. Science 2013;340(6132) :630–5. 32. Stemberger C, Huster K, Koffler M, Anderl F, Schiemann M, Wagner H, et al. A single naive CD8+ T cell precursor can develop into diverse effector and memory subsets. Immunity 2007;27(6):985–97. 33. Stemberger C, Graef P, Odendahl M, Albrecht J, Dossinger G, Anderl F, et al. Lowest numbers of primary CD8(+) T cells can reconstitute protective immunity upon adoptive immunotherapy. Blood 2014;124(4):628–37. 34. Levine BL, Mosca J, Riley JL, Carroll RG, Vahey MT, Jagodizinski L, et al. Antiviral effect and ex vivo CD4+ T cell proliferation in HIV-positive patients as a result of CD28 costimulation. Science 1996;272:1939–43. 35. Levine BL, Bernstein W, Craighead N, Lindsten T, Thompson CB, June CH. Effects of CD28 costimulation on long term proliferation of CD4+ T cells in the absence of exogenous feeder cells. J Immunol 1997;159(12):5921–30. 36. Levine BL, Bernstein WB, Aronson NE, Schlienger K, Cotte J, Perfetto S, et al. Adoptive transfer of costimulated CD4+ T cells induces expansion of peripheral T Cells and decreased CCR5 expression in HIV infection. Nat Med 2002;8(1):47–53. 37. Scholler J, Brady T, Binder-Scholl G, Hwang W-T, Plesa G, Hege K, et al. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci Transl Med 2012;4(132) 132Ra53. 38. Tebas P, Stein D, Tang W, Frank I, Wang S, Lee G, et al. Gene editing of CCR5 in autologous CD4 T-cells of persons infected with HIV. N Engl J Med 2014;370(10):901–10. 39. Quezada SA, Peggs KS, Simpson TR, Allison JP. Shifting the equilibrium in cancer immunoediting: from tumor tolerance to eradication. Immunol Rev 2011;241(1):104–18. 40. Silver PA, Way JC, Arnold FH, Meyerowitz JT. Synthetic biology: engineering explored. Nature 2014;509(7499):166–7. 41. Way JC, Collins JJ, Keasling JD, Silver PA. Integrating biological redesign: where synthetic biology came from and where it needs to go. Cell 2014;157(1):151–61. 42. Wu CY, Roybal KT, Puchner EM, Onuffer J, Lim WA. Remote control of therapeutic T cells through a small molecule-gated chimeric receptor. Science 2015;350(6258):aab4077. 43. Rufer N, Migliaccio M, Antonchuk J, Humphries RK, Roosnek E, Lansdorp PM. Transfer of the human telomerase reverse transcriptase (TERT) gene into T lymphocytes results in extension of replicative potential. Blood 2001;98(3):597–603.

Adoptive Cellular Therapy Chapter | 30

593

44. Eaton D, Gilham DE, O’Neill A, Hawkins RE. Retroviral transduction of human peripheral blood lymphocytes with Bcl-X(L) promotes in vitro lymphocyte survival in pro-apoptotic conditions. Gene Ther 2002;9(8):527–35. 45. Dotti G, Savoldo B, Pule M, Straathof KC, Biagi E, Yvon E, et al. Human cytotoxic T lymphocytes with reduced sensitivity to Fas-induced apoptosis. Blood 2005;105(12):4677–84. 46. Bollard CM, Rossig C, Calonge MJ, Huls MH, Wagner HJ, Massague J, et al. Adapting a transforming growth factor beta-related tumor protection strategy to enhance antitumor immunity. Blood 2002;99(9):3179–87. 47. Gaj T, Gersbach CA, Barbas III CF. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol 2013;31(7):397–405. 48. 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(7486):52–7. 49. Parnas O, Jovanovic M, Eisenhaure TM, Herbst RH, Dixit A, Ye CJ, et al. A genome-wide CRISPR screen in primary immune cells to dissect regulatory networks. Cell 2015;162(3):675–86. 50. Konermann S, Brigham MD, Trevino AE, Joung J, Abudayyeh OO, Barcena C, et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 2015;517(7536):583–8. 51. Naldini L. Ex vivo gene transfer and correction for cell-based therapies. Nat Rev Genet 2011;12(5):301–15. 52. Suerth JD, Schambach A, Baum C. Genetic modification of lymphocytes by retrovirus-based vectors. Curr Opin Immunol 2012;24(5):598–608. 53. Hackett PB, Aronovich EL, Hunter D, Urness M, Bell JB, Kass SJ, et al. Efficacy and safety of sleeping beauty transposon-mediated gene transfer in preclinical animal studies. Curr Gene Ther 2011;11:341–9. 54. Piganeau M, Ghezraoui H, De Cian A, Guittat L, Tomishima M, Perrouault L, et al. Cancer translocations in human cells induced by zinc finger and TALE nucleases. Genome Res 2013;23(7):1182–93. 55. Berdien B, Mock U, Atanackovic D, Fehse B. TALEN-mediated editing of endogenous T-cell receptors facilitates efficient reprogramming of T lymphocytes by lentiviral gene transfer. Gene Ther 2014;21(6):539–48. 56. Poirot L, Philip B, Schiffer-Mannioui C, Le Clerre D, Chion-Sotinel I, Derniame S, et al. Multiplex genome-edited T-cell manufacturing platform for “off-the-shelf” adoptive T-cell immunotherapies. Cancer Res 2015;75(18):3853–64. 57. Schumann K, Lin S, Boyer E, Simeonov DR, Subramaniam M, Gate RE, et al. Generation of knock-in primary human T cells using Cas9 ribonucleoproteins. Proc Natl Acad Sci USA 2015;112(33):10437–42. 58. Kuwana Y, Asakura Y, Utsunomiya N, Nakanishi M, Arata Y, Itoh S, et al. Expression of chimeric receptor composed of immunoglobulin-derived V regions and T-cell receptor-derived C regions. Biochem Biophys Res Commun 1987;149(3):960–8. 59. 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(24):10024–8. 60. Brown CE, Badie B, Barish ME, Weng L, Ostberg JR, Chang W-C, et al. Bioactivity and safety of IL13Rα2-redirected chimeric antigen receptor CD8+ T cells in patients with recurrent glioblastoma. Clin Cancer Res 2015;21:4062–72. 61. Johnson LA, Scholler J, Ohkuri T, Kosaka A, Patel PR, McGettigan SE, et al. Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci Transl Med 2015;7(275):275ra22.

594

PART | XI  Future Challenges

62. Hudecek M, Lupo-Stanghellini MT, Kosasih PL, Sommermeyer D, Jensen MC, Rader C, et al. Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1specific chimeric antigen receptor T cells. Clin Cancer Res 2013;19(12):3153–64. 63. Liu X, Jiang S, Fang C, Yang S, Olalere D, Pequignot EC, et al. Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice. Cancer Res 2015;75(17):3596–607. 64. Moritz D, Groner B. A spacer region between the single-chain antibody-chain and the CD3 zeta-chain domain of chimeric T-cell receptor components is required for efficient ligand-binding and signaling activity. Gene Ther 1995;2002:539–46. 65. Hombach A, Hombach A, Abken H. Adoptive immunotherapy with genetically engineered T cells: modification of the IgG1 Fc “spacer” domain in the extracellular moiety of chimeric antigen receptors avoids “off-target” activation and unintended initiation of an innate immune response. Gene Ther 2010;17:1206–13. 66. Rosenberg SA, Terry WD. Passive immunotherapy of cancer in animals and man. Adv Cancer Res 1977;25:323–88. 67. Walter EA, Greenberg PD, Gilbert MJ, Finch RJ, Watanabe KS, Thomas ED, et al. Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor. N Engl J Med 1995;333(16):1038–44. 68. Heslop HE, Ng CY, Li C, Smith CA, Loftin SK, Krance RA, et al. Long-term restoration of immunity against Epstein–Barr virus infection by adoptive transfer of gene-modified virusspecific T lymphocytes. Nat Med 1996;2(5):551–5. 69. Kiepiela P, Ngumbela K, Thobakgale C, Ramduth D, Honeyborne I, Moodley E, et al. CD8(+) T-cell responses to different HIV proteins have discordant associations with viral load. Nat Med 2007;13(1):46–53. 70. Brodie SJ, Patterson BK, Lewinsohn DA, Diem K, Spach D, Greenberg PD, et al. HIV-specific cytotoxic T lymphocytes traffic to lymph nodes and localize at sites of HIV replication and cell death. J Clin Invest 2000;105(10):1407–17. 71. Koenig S, Conley AJ, Brewah YA, Jones GM, Leath S, Boots LJ, et al. Transfer of HIV-1-specific cytotoxic T lymphocytes to an AIDS patient leads to selection for mutant HIV variants and subsequent disease progression. Nat Med 1995;1(4):330–6. 72. Lund O, Lund OS, Gram G, Nielsen SD, Schonning K, Nielsen JO, et al. Gene therapy of T helper cells in HIV infection: mathematical model of the criteria for clinical effect. Bull Math Biol 1997;59(4):725–45. 73. Wodarz D, Nowak MA. Specific therapy regimes could lead to long-term immunological control of HIV. Proc Natl Acad Sci USA 1999;96(25):14464–9. 74. Varela-Rohena A, Molloy PE, Dunn SM, Li Y, Suhoski MM, Carroll RG, et al. Control of HIV-1 immune escape by CD8 T cells expressing enhanced T-cell receptor. Nat Med 2008;14(12):1390–5. 75. June C, Levine B. Blocking HIV’s attack. Sci Am 2012;306(3):54–9. 76. 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(6):747–52. 77. Kochenderfer J, Wilson W, Janik J, Dudley M, Stetler-Stevenson M, Feldman S, 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(20):4099–102. 78. Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol 2011;29(7):917.

Adoptive Cellular Therapy Chapter | 30

595

79. Rapoport AP, Stadtmauer EA, Binder-Scholl GK, Goloubeva O, Vogl DT, Lacey SF, et al. NYESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat Med 2015;21(8):914–21. 80. Porter DL, Roth MS, McGarigle C, Ferrara JL, Antin JH. Induction of graft-versus-host disease as immunotherapy for relapsed chronic myeloid leukemia. N Engl J Med 1994;330:100–6. 81. Bluestone JA, Buckner JH, Fitch M, Gitelman SE, Gupta S, Hellerstein MK, et al. Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci Transl Med 2015;7(315):315ra189. 82. Juvet S, Whatcott A, Bushell A, Wood K. Harnessing regulatory T cells for clinical use in transplantation: the end of the beginning. Am J Transplant 2014;14(4):750–63. 83. Brunstein CG, Miller JS, McKenna DH, Hippen KL, DeFor TE, Sumstad D, et al. Umbilical cord blood-derived T regulatory cells to prevent GVHD: kinetics, toxicity profile and clinical effect. Blood 2016;127(8):1044–51. 84. Riddell SR, Watanabe KS, Goodrich JM, Li CR, Agha ME, Greenberg PD. Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science 1992;257:238–41. 85. Mitsuyasu RT, Anton P, Deeks SG, Scadden DT, Connick E, Downs MT, et al. Prolonged survival and tissue trafficking following adoptive transfer of CD4 z gene-modified autologous CD4+ and CD8+ T cells in HIV-infected subjects. Blood 2000;96(3):785–93. 86. Maus MV, Grupp SA, Porter DL, June CH. Antibody modified T cells: CARs take the front seat for hematologic malignancies. Blood 2014;123(17):2625–35. 87. Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res 2006;12(20 Pt. 1):6106–15. 88. Lamers CH, Sleijfer S, Vulto AG, Kruit WH, Kliffen M, Debets R, et al. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol 2006;24(13):e20–2. 89. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 2011;365(8):725–33. 90. Brentjens R, Rivière I, Park J, Davila M, Wang X, Stefanski J, et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 2011;118(18):4817–28. 91. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter D, Rheingold S, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 2013;368(16):1509–18. 92. Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 2015;385(9967):517–28. 93. Tedder TF. CD19: a promising B cell target for rheumatoid arthritis. Nat Rev Rheumatol 2009;5(10):572–7. 94. Maude S, Frey N, Shaw P, Aplenc R, Barrett D, Bunin N, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 2014;371(16):1507–17. 95. Garfall AL, Maus MV, Hwang WT, Lacey SF, Mahnke YD, Melenhorst JJ, et al. Chimeric antigen receptor T cells against CD19 for multiple myeloma. N Engl J Med 2015;373(11):1040–7. 96. Porter DL, Hwang WT, Frey NV, Lacey SF, Shaw PA, Loren AW, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med 2015;7(303):303ra139. 97. 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(177):177ra38.

596

PART | XI  Future Challenges

98. Sotillo E, Barrett DM, Black KL, Bagashev A, Oldridge D, Wu G, et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov 2015;5:1282–95. 99. Mukherjee S. The emperor of all maladies: a biography of cancer. New York, NY: Simon and Schuster; [2011] 100. Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, et al. T cells expressing chimeric receptors establish memory and potent antitumor effects in patients with advanced leukemia. S Trans Med 2011;3(95):95ra73. 101. Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, et al. How I treat: current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014;124(2):188–95. 102. Barrett DM, Teachey DT, Grupp SA. Toxicity management for patients receiving novel T-cell engaging therapies. Curr opin Pediatrics 2014;26(1):43–59. 103. Fitzgerald J, Weiss SL, Maude SL, Barrett DM, Lacey SF, Melenhorst JJ, et al. Cytokine release syndrome after chimeric antigen receptor T Cell Therapy for acute lymphoblastic leukemia. Crit Care Med 2016; [in press]. 104. Topp MS, Kufer P, Gokbuget N, Goebeler M, Klinger M, Neumann S, et al. Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J Clin Oncol 2011;29(18):2493–8. 105. Levine BL, June CH. Perspective: assembly line immunotherapy. Nature 2013;498(7455):S17. 106. Torikai H, Reik A, Liu P-Q, Zhou Y, Zhang L, Maiti S, et al. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood 2012;119(24):5697–705. 107. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015;348(6230):62–8.