Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Apr 25.
Published in final edited form as: Clin Lymphoma Myeloma Leuk. 2014 Sep;14(Suppl):S18–S22. doi: 10.1016/j.clml.2014.06.013

Progress in Novel Cellular Therapy Options for CLL: The MD Anderson Perspective

Nina Shah 1, Katy Rezvani 1, Chitra Hosing 1, Partow Kebriaei 1, William Wierda 2, Laurence Cooper 3, Elizabeth Shpall 1
PMCID: PMC4844462  NIHMSID: NIHMS773726  PMID: 25486950

Abstract

Over the past several years there has been considerable progress in the number and breadth of therapeutic options for patients with chronic lymphocytic leukemia (CLL). In particular, cumulative experience with stem cell transplantation and immunotherapy has made these modalities more available to a broader range of patients. Advances in genetic engineering and ex vivo expansion techniques have sculpted cellular therapy products to optimize the combination of specificity and toxicity. In this brief review, we will discuss recent cutting-edge experience with chimeric antigen receptor T cells as well as developing cellular therapy products at our institution.

INTRODUCTION

Chronic lymphocytic leukemia (CLL) is the most common adult hematologic malignancy. While some patients can demonstrate a long, indolent course without significant morbidity, a significant number of patients eventually require treatment. Though there has been a remarkable development in the treatment options for CLL over the past 15 years, many patients, particularly those with high-risk features, often succumb to their disease.

Allogeneic hematopoietic stem cell transplantation (HSCT) has emerged as a viable treatment strategy for CLL patients and is still considered the only curative option. Advances in preparative regimens have now made this procedure more tolerable for older patients. In addition, reduced intensity regimens underscore the importance of the graft versus leukemia (GVL) effect in CLL. Though this regime has been highly successful with some centers reporting 39–70% long term survival,1 the issue of relapse still remains, as does minimizing graft versus host disease (GVHD) while maximizing GVL.

Building on the beneficial effect of GVL seen in CLL, numerous investigators have been developing strategies to optimize cell therapy to yield a more durable and eventually less toxic approach to eradicating this disease. In this brief report we will discuss the results of early novel cell therapy trials in CLL as well as unveil new strategies being developed at our institution and elsewhere.

CHIMERIC ANTIGEN RECEPTOR T CELLS

One of the most highly publicized and exciting developments over the past few years has been engineering of T cells to express chimeric antigen receptors (CARs). The CAR technology combines the specificity of antibody therapy with the cytotoxicity of T cells to directly target tumor cells. Each CAR is expressed on a T cell as a result of a genetic integration of a specific construct with the native DNA.

The basic design of a CAR involves an extracellular domain which is antigen-specific and an intracellular signaling domain which prompts cytotoxicity. The antigen specific extracellular domain is most commonly derived from the variable region of the respective antibody produced by B-cells. Thus, recognition of an antigen by these CAR T cells does not depend on presentation by HLA molecules. In the case of CLL, CD19 has been the tumor associated antigen (TAA) of choice, as it is essentially limited to cells of B-lineage. The signaling portion of the CAR relies on machinery more native to the T cell receptor, though the optimal sequence of signaling domains is still evolving. The more recent modifications of this technology have employed the CD3ζ activation domain combined with that of one or more co-stimulatory receptors such as CD28 and 4-1BB. The transduction of T cells by the CAR genetic construct is accomplished by various means, including retroviral vectors and transposases.

Thus far there have been several clinical trials of CAR T cells that have included patients with CLL. The majority of these are early phase studies to establish safety and feasibility of this technology. In 2011 Kalos et al reported a pilot trial of 3 patients with advanced CLL who were treated with autologous CD19-specific CAR T cells with the CD3ζ/4-1BB construct.2 Overall 2 patients achieved a complete response (CR) and 1 patient achieved a partial response (PR). An recent update of these results in abstract format demonstrated responses in 8/14 CLL patients.3 In addition, an ongoing dose-optimizing study from the same institution has demonstrated a 40% response rate in relapsed/refractory CLL patients.4 A similar trial conducted by Kochenderfer et al enrolled 8 patients with B- cell malignancies, 4 of whom had CLL.5 Patients received CD19-specific CAR T cells (CD3ζ/CD28 endodomain) and supplemental IL-2. Of the 4 CLL patients included in this trial 1 achieved a CR, 2 achieved a PR and 1 patient had stable disease (SD). Brentjens et al treated 8 patients with CD19-specific CAR T cells (CD3ζ/CD28 endodomain) and reported 3 patients with SD.6 Importantly, this study demonstrated trafficking of T cells to tumor sites and persistence up to 8 days after infusion. The toxicity in these studies has varied; several patients have experienced hypotension and other signs of cytokine release storm (CRS). Preliminarily, this CRS may actually be a surrogate for clinical response.7

More recently, there have been clinical trials employing allogeneic CAR T cells. Kochenderfer et al treated 10 allogeneic HSCT recipients with donor-derived CD19-specific CAR T cells.8 Of the 4 CLL patients included in this trial, 1 achieved a CR and another maintained SD without additional GVHD. Similarly, the group at Baylor College of Medicine has demonstrated the ability to transduce virus-specific allogenenic T-cells with CD19-specific CAR.9 This study included 4 patients with CLL who had relapsed after allogeneic HSCT. Of these 4 patients, 1 achieved a PR and another achieved SD. Importantly, there were no infusion-related toxicities and no associated GVHD. These studies thus presented a new platform for treating CLL patients with allogeneic CD19-specific T cell therapy, opening the door for patients who relapse after allogeneic HSCT or as a part of early post-HSCT consolidation.

At our institution we are building on the growing knowledge in this field to offer patients both autologous and allogeneic CAR T cell products. Because of the increased flexibility in rapidly developing new constructs we are exploring the Sleeping Beauty transposon/transposase system.10 This system has the dual advantage of stable introduction of the CD19-specific CAR sequence and deletion of the α or β T cell receptor (TCR) chains, thus limiting unwanted, non-specific allo-reactivity. With this technology we have been able to generate CD19-specific T cells which are not responsive to TCR stimulation. Using an artificial antigen presenting cell (APC) culture system, we have been able to expand these CAR T cells >1000-fold. We are currently conducting 4 clinical trials of either autologous or allogeneic CD19-specific CAR T cells: 2 trials for B cell malignancies,11 a cord blood-CAR-CD19 trial and a trial of autologous CAR T cells specifically for CLL patients in the non-transplant setting. To date there have been no significant toxicities in any of these trials while efficacy data is still being matured. In select patients, the CAR T cells have been detectable several months post-infusion.

While CD19 is an attractive target for CAR technology, additional TAAs are being studied. In collaboration with other institutions we are also developing a CAR against receptor tyrosine kinase-like orphan receptor-1 (ROR1). This protein is expressed highly on CLL cells as well as mantle cell lymphoma cells with some additional, lover level expression on adipose tissue.12 Importantly, ROR1 is not expressed on mature B cells. This target thus has the potential advantage of malignant B-cell specificity without total depletion of all B cell stores. In addition ROR1 may also be expressed on solid tumor cells.13 Though there is the potential for low level ROR1 expression on normal tissue cells, there are ROR1 antibodies in development that do not bind to normal tissues. In preliminarily studies, we have been able to transduce normal donor T cells with a ROR1-specific CAR (containing either a CD3ζ/CD28 or CD3ζ/4-1BB endodomain) with subsequent logarithmic expansion of T cells. These cells demonstrate in vivo activity in a murine model of B cell malignancy.

NATURAL KILLER CELLS

Natural Killer (NK) cells are increasingly becoming recognized as important players in innate anti-tumor immunity. These CD56+/CD3 lymphocytes are thought to become activated after engagement with cells which lack MHC class I (the “missing-self hypothesis”).14 They can thus recognize tumor cells which tend to down regulate MHC class I. Furthermore, in an allogeneic HSCT setting, certain mismatches between donor-recipient HLA alleles are associated with theoretical NK cell alloreactivity and improved clinical outcome without concomitant increases in GVHD.15,16 In addition, NK cell recovery after allogeneic HSCT has been linked to decreased acute GVHD, decreased non-relapse mortality and improved relapse-free survival.17 To date the majority of clinical trials with NK cell therapy have focused on myeloid malignancies but there is a growing interest in the potential application of NK cells against CLL.

While autologous NK cell generation from CLL patients would seem attractive, this is challenging due to several factors. The absolute number of NK cells from CLL patients is limited, particularly with the relative increase in B cells, thus impairing the generation of a clinically meaningful NK cell dose. In addition, there is growing evidence to suggest that autologous NK cells from CLL patients are hypofunctional with decreased NKp30 expression.18

Though the field of NK cell therapy has traditionally relied on peripheral blood-derived NK cells we have been interested in harvesting NK cells from umbilical cord blood (CB), a known source of hematopoietic stem cells. Recently our group has developed a system for large-scale, GMP-compliant ex vivo expansion of CB-derived NK cells from a fraction of a frozen CB unit.19 These cells demonstrate an activated phenotype and are cytotoxic against K562 cells as well as multiple myeloma cells.

Preliminary data from our laboratory suggests that CB-derived NK cells are also active against CLL. After ex vivo expansion, CB-NK cells appear to form strong synapses with target primary CLL cells and demonstrate in vitro cytotoxicity (Figure 1A and B). In a murine model of CLL that is under development in our lab, these CB-NK cells appear to impair in vivo expansion of CLL cells and subsequent lymphoid infiltration of the spleen (Figure 2A and B).

Figure 1.

Figure 1

A. Ex vivo expanded CB-NK cells form immunological synapses with primary CLL cells. B. CB-NK cells demonstrate dose-dependent cytotoxicity against primary CLL cells in a standard Chromium-51 assay.

Figure 2.

Figure 2

CFSE-labeled CLL cells were injected into NOD/SCID IL-2Rγnull (NSG) mice with our without CB-NK cell adoptive transfer. A. Representative plot at day 12 demonstrating impaired proliferation of CFSE-labeled CLL cells in the mice who received CB-NK cells. B. Hematoxylin and eosin staining showing reduced lymphocytic infiltration of murine spleen after treatment with CB-NK cells.

There is also emerging data from our laboratory and others’20,21 suggesting that NK cell function may be augmented by lenalidomide. Our preliminary findings demonstrate increased in vitro CB-NK cytotoxicity against CLL after incubation with lenalidomide (Figure 3). Though the mechanisms underlying this interaction are not clear, multiple hypotheses have been proposed, including disinhibition of NK cell signaling,22 increase in NK cell activating receptors23 and enhancement of NK-CLL synapse formation.24 These results suggest that the eventual combination of immunomodulatory and cellular therapies may optimize immune therapy for CLL.

Figure 3.

Figure 3

Chromium-51 assay demonstrating augmented CB-NK cytotoxicity against primary CLL cells in the presence of lenalidomide.

Finally, we have recently transduced CB-NK cells with a CD19-retroviral CAR construct that also expresses IL15. In preliminary experiments these NK-CAR-CD19 cells have shown significantly higher levels of cytotoxicity against CD19+ target cells than non-transduced NK cells. Once confirmed, translation of NK-CAR therapy to the clinic is planned.

CONCLUSION

In conclusion, the therapeutic options for patients with CLL have increased dramatically in the last several years. While there has been an unprecedented development in pharmacologic agents, there has been an equally exciting progress in cellular therapy. The multiple clinical trials and modifications of CAR T cells are beginning to converge on the optimal strategy for this technology. In addition, the advances in ex vivo expansion and immunomodulation have brought within reach NK cell therapy. As we continue to improve specificity and limit toxicity, we can move closer to the ideal goal of long-term remission for our patients.

Acknowledgments

Funding Sources: CLL Global Research Foundation: Alliance Program

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of Interest: Dr. Cooper founded and owns InCellerate, Inc.; InCellerate is not involved in the development of CAR T cells. He has patents with Sangamo BioSciences with artificial nucleases. He consults with Targazyme, Inc. (American Stem cells, Inc.), GE Healthcare, Ferring Pharmaceuticals, Inc., and Bristol-Myers Squibb.

References

  • 1.Kharfan-Dabaja MA, Bazarbachi A. Hematopoietic stem cell allografting for chronic lymphocytic leukemia: a focus on reduced-intensity conditioning regimens. Cancer control: journal of the Moffitt Cancer Center. 2012;19(1):68–75. doi: 10.1177/107327481201900107. [DOI] [PubMed] [Google Scholar]
  • 2.Kalos M, Levine BL, Porter DL, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Science translational medicine. 2011;3(95):95ra73. doi: 10.1126/scitranslmed.3002842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Porter D, Kalos M, Frey N, et al. Chimeric Antigen Receptor Modified T Cells Directed Against CD19 (CTL019 cells) Have Long-Term Persistence and Induce Durable Responses In Relapsed, Refractory CLL. American Society of Hematology Annual Meeting. 2013 Abstract 4162. [Google Scholar]
  • 4.Porter D, Kalos M, Frey N, et al. Randomized, Phase II Dose Optimization Study Of Chimeric Antigen Receptor Modified T Cells Directed Against CD19 (CTL019) In Patients With Relapsed, Refractory CLL. American Society of Hematology Annual Meeting. 2013 Abstract 873. [Google Scholar]
  • 5.Kochenderfer JN, Dudley ME, Feldman SA, 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(12):2709–2720. doi: 10.1182/blood-2011-10-384388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Brentjens RJ, Riviere I, Park JH, 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):4817–4828. doi: 10.1182/blood-2011-04-348540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kalos M, Nazimuddin F, Finklestein J, et al. Long-Term Functional Persistence, B Cell Aplasia Anti-Leukemia Efficacy In Refractory B Cell Malignancies Following T Cell Immunotherapy Using CAR-Redirected T Cells Targeting CD19. American Society of Hematology Annual Meeting. 2013 Abstract 163. [Google Scholar]
  • 8.Kochenderfer JN, Dudley ME, Carpenter RO, et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood. 2013;122(25):4129–4139. doi: 10.1182/blood-2013-08-519413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Cruz CR, Micklethwaite KP, Savoldo B, et al. Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study. Blood. 2013;122(17):2965–2973. doi: 10.1182/blood-2013-06-506741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Singh H, Figliola MJ, Dawson MJ, et al. Manufacture of clinical-grade CD19-specific T cells stably expressing chimeric antigen receptor using Sleeping Beauty system and artificial antigen presenting cells. PloS one. 2013;8(5):e64138. doi: 10.1371/journal.pone.0064138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kebriaei P, Huls H, Jena B, et al. Infusing CD19-directed T cells to augment disease control in patients undergoing autologous hematopoietic stem-cell transplantation for advanced B-lymphoid malignancies. Human gene therapy. 2012;23(5):444–450. doi: 10.1089/hum.2011.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hudecek M, Schmitt TM, Baskar S, et al. The B-cell tumor-associated antigen ROR1 can be targeted with T cells modified to express a ROR1-specific chimeric antigen receptor. Blood. 2010;116(22):4532–4541. doi: 10.1182/blood-2010-05-283309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Rabbani H, Ostadkarampour M, Danesh Manesh AH, Basiri A, Jeddi-Tehrani M, Forouzesh F. Expression of ROR1 in patients with renal cancer–a potential diagnostic marker. Iranian biomedical journal. 2010;14(3):77–82. [PMC free article] [PubMed] [Google Scholar]
  • 14.Passweg JR, Stern M, Koehl U, Uharek L, Tichelli A. Use of natural killer cells in hematopoetic stem cell transplantation. Bone marrow transplantation. 2005;35(7):637–643. doi: 10.1038/sj.bmt.1704810. [DOI] [PubMed] [Google Scholar]
  • 15.Ruggeri L, Capanni M, Mancusi A, et al. Alloreactive natural killer cells in mismatched hematopoietic stem cell transplantation. Blood cells, molecules & diseases. 2004;33(3):216–221. doi: 10.1016/j.bcmd.2004.08.005. [DOI] [PubMed] [Google Scholar]
  • 16.Hsu KC, Gooley T, Malkki M, et al. KIR ligands and prediction of relapse after unrelated donor hematopoietic cell transplantation for hematologic malignancy. Biology of blood and marrow transplantation: journal of the American Society for Blood and Marrow Transplantation. 2006;12(8):828–836. doi: 10.1016/j.bbmt.2006.04.008. [DOI] [PubMed] [Google Scholar]
  • 17.Savani BN, Rezvani K, Mielke S, et al. Factors associated with early molecular remission after T cell-depleted allogeneic stem cell transplantation for chronic myelogenous leukemia. Blood. 2006;107(4):1688–1695. doi: 10.1182/blood-2005-05-1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Veuillen C, Aurran-Schleinitz T, Castellano R, et al. Primary B-CLL resistance to NK cell cytotoxicity can be overcome in vitro and in vivo by priming NK cells and monoclonal antibody therapy. Journal of clinical immunology. 2012;32(3):632–646. doi: 10.1007/s10875-011-9624-5. [DOI] [PubMed] [Google Scholar]
  • 19.Shah N, Martin-Antonio B, Yang H, et al. Antigen presenting cell-mediated expansion of human umbilical cord blood yields log-scale expansion of natural killer cells with anti-myeloma activity. PloS one. 2013;8(10):e76781. doi: 10.1371/journal.pone.0076781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Zhu D, Corral LG, Fleming YW, Stein B. Immunomodulatory drugs Revlimid (lenalidomide) and CC-4047 induce apoptosis of both hematological and solid tumor cells through NK cell activation. Cancer immunology, immunotherapy: CII. 2008;57(12):1849–1859. doi: 10.1007/s00262-008-0512-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hsu AK, Quach H, Tai T, et al. The immunostimulatory effect of lenalidomide on NK-cell function is profoundly inhibited by concurrent dexamethasone therapy. Blood. 2011;117(5):1605–1613. doi: 10.1182/blood-2010-04-278432. [DOI] [PubMed] [Google Scholar]
  • 22.Gorgun G, Calabrese E, Soydan E, et al. Immunomodulatory effects of lenalidomide and pomalidomide on interaction of tumor and bone marrow accessory cells in multiple myeloma. Blood. 2010;116(17):3227–3237. doi: 10.1182/blood-2010-04-279893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lioznov M, El-Cheikh J, Jr, Hoffmann F, et al. Lenalidomide as salvage therapy after allo-SCT for multiple myeloma is effective and leads to an increase of activated NK (NKp44(+)) and T (HLA-DR(+)) cells. Bone marrow transplantation. 2010;45(2):349–353. doi: 10.1038/bmt.2009.155. [DOI] [PubMed] [Google Scholar]
  • 24.Gaidarova S, Li J, Corral L, et al. Lenalidomide Alone and in Combination with Rituximab Enhances NK Cell Immune Synapse Formation in Chronic Lymphocytic Leukemia (CLL) Cells in Vitro through Activation of Rho and Rac1 GTPases. American Society of Hematology Annual Meeting. 2009 Abstract 3441. [Google Scholar]

RESOURCES