Loading

Journal of Cancer Biology and Research

Chimeric Antigen Receptor Therapy as a Means of Cellular Immunotherapy in B Cell Malignancies

Review Article | Open Access | Volume 5 | Issue 3

  • 1. Department of Medicine, Vanderbilt University Medical Center, USA
+ Show More - Show Less
Corresponding Authors
Olalekan O. Oluwole, Hematology Oncology Division, Department of Medicine, Vanderbilt University Medical Center, 1301 Medical Center Drive (3927 TVC), Nashville TN, 37232-5505, USA, Tel: 615 9368422
ABSTRACT

Chimeric antigen receptor T cells (CARs) are a form of cellular immunotherapy whereby a  patient’s T cells are re-engineered to express a chimeric tumor antigen-specific T cell receptor,  leading to activation and expansion upon recognition of target antigen-bearing cells. Some  patients with B cell malignancies, chemotherapy may have limited efficacy in achieving the  disease control necessary for undergoing hematopoietic stem cell transplant, which is often  the only curative option. However, numerous phase 1 and ongoing phase 2 clinical trials have  demonstrated the efficacy of CD19-targeted CARs at achieving disease control in relapsed and  refractory patients in a range of B cell malignancies, with response rates as high as 60-90%.  Side effects associated with CAR therapy may be severe but are manageable and include  cytokine release syndrome, neurotoxicity, and B cell aplasia. In this article, we present a review  of the CAR construct, early and ongoing CAR trials in B cell malignancies, the presentation,  monitoring, and management of major side effects associated with CAR therapy, and future  directions for CAR research and development.

KEYWORDS

Chimeric antigen receptor;Human leukocyte antigen ;Immunotherapy ; Chronic lymphocytic leukemia ;Tumor necrosis factor ;Ribonucleic acid.

CITATION

Wright FM, Oluwole OO (2017) Chimeric Antigen Receptor Therapy as a Means of Cellular Immunotherapy in B Cell Malignancies. J Cancer Biol Res 5(3): 1102

ABBREVIATIONS

CAR: Chimeric Antigen Receptor; FDA: Food and Drug Administration; APC: Antigen Presenting Cell; HLA: Human Leukocyte Antigen; CLL: Chronic Lymphocytic Leukemia; ALL: Acute Lymphoblastic Leukemia; MRD: Minimal Residual Disease; DLBCL: Diffuse Large B Cell Lymphoma; CRS: Cytokine Release Syndrome; CRP: C Reactive Protein; TNF: Tumor Necrosis Factor; MAS: Macrophage Activation Syndrome; HLH: Hemophagocytic Lymphohistiocytosis; DLBCL: Diffuse Large B Cell Lymphoma; IVIG: Intravenous Immunoglobulin’s; EEG: Electroencephalogram; GVHD: Graft Versus Host Disease; LP: Lumbar Puncture; HLA: Human Leukocyte Antigen; CD: Cluster of Differentiation; IL: Interleukin; CNS: Central Nervous System; RNA: Ribonucleic Acid; DNA: Deoxyribonucleic Acid; PD-1: Programmed Cell Death Protein 1

INTRODUCTION

Cellular immunotherapy in the treatment of B-cell malignancies arose because existing salvage chemotherapy options are of limited efficacyand are associated with significant morbidity and mortality, including severe or prolonged myelosuppression, cardiac toxicity, and other organ dysfunction [1]. Patients with relapsed B-cell malignancies who have chemotherapy-sensitive disease are often required to undergo autologous, and in some cases, allogeneic stem cell transplant for disease control [2,3]. However, only a fraction of patients ultimately qualify for transplant, either because their disease is not sufficiently chemotherapy-sensitive, or because they have suffered complications with salvage chemotherapy that preclude further treatment. Prognosis in either instance is poor. On the other hand, patients who are able to qualify for and undergo allogeneic stem cell transplant, for example, for acute lymphoblastic leukemia, have significant disease-free and overall survival compared to salvage chemotherapy alone [4,5]. For many patients, the primary barrier to transplant is inadequate disease control. The success of CAR trials in the setting of B-cell malignancies, including their role as a bridge to transplant, represents a major advance for immunotherapy in the treatment and eventual eradication of select B cell malignancies. In this review, we present an overview of cellular immunotherapy using chimeric antigen receptor (CAR) technology. We also present some of the recently concluded clinical trials and enumerate ongoing trials. Lastly, we hypothesize about the future of CAR therapy, and cellular immunotherapy in general, in the treatment of hematologic malignancies.

THE CAR CONSTRUCT

The part-human, part-murine chimeric antigen receptor construct is analogous to a monoclonal antibody attached to the intracellular signaling domain of a T cell receptor. It is designed to initiate intracellular signaling upon binding its target cell surface antigen, precipitating T cell activation and clonal expansion in a manner that is independent of Human Leukocyte Antigen (HLA) presentation [6,7]. This design allows for target selection according to tumor cell antigen expression and is capable of inducing a potent antigen-specific immune response.

Several experiments in the 1980s and 1990s paved the way for the development of the essential components of the CAR construct [8,9]. In order to make the CAR construct, a single-stranded DNA fragment is delivered into the mature T lymphocyte using a gamma retro viral or lent viral vector. The CAR transgene comprises a single chain variable fragment (scFv) for antigen recognition, the CD3 zeta chain (CD3z) derived from the T cell intracellular signaling cascade, and at least one costimulatory domain (typically, CD28z or 4-1BBz) [10-12]. The scFv is, in effect, a synthetic monoclonal antibody; it is composed of the variable portions of the heavy and light chains linked by a hinge domain (Figure 1).

1 Components of the CAR construct

Figure 1 Components of the CAR construct.

EARLY CLINICAL DATA

The first generation CARs, consisting of only anscFvecto domain linked to a CD3z endo domain, were able to bind to the specified target, but they were unable to multiply and persist in the host sufficiently to mount a sustained and effective immune response. The CAR construct was intended to render T cell activation independent of antigen presenting cells (APC), but these early CARs importantly lacked the costimulatory signal normally generated through interaction of the activated T cell and the APC (CD28 on the T cell and CD80/86 on the APC) necessary for a robust and sustained immune response. Thus, second generation CARs incorporated a costimulatory domain (CD28z or 4-1BBz) into the CAR endo domain in sequence with the CD3z. These CARs were able to mount a robust antitumor response and persisted to ensure successful eradication of the defined target, including malignant cells, and they resulted in clinically meaningful response rates. Preclinical studies suggested even greater antitumor activity with “third-generation” CARs, which incorporate an additional costimulatory domain in sequence with the CD3z and CD28z signaling moieties [6,13]. Ultimately, however, they were not proven to convey increased efficacy,and thus most current trials employ second generation CARs [14]. An illustration of the first, second and third generation CAR constructs is shown in Figure 2.

2 Schematic Depiction of Chimeric Antigen Receptor

Figure 2 Schematic Depiction of Chimeric Antigen Receptor.

SELECT COMPLETED TRIALS USING CAR T THERAPY

Several non-randomized, mostly small phase 1 and singleinstitution clinical trials have provided valuable insights into the efficacy and safety of CAR therapy. These trials have also informed strategies to predictably enhance as well as control CARs, leading to the successful development of single arm phase 2 clinical trials.

Chronic lymphocytic leukemia (CLL)

One of the earliest human experiments using CAR therapy was performed in patients with relapsed, refractory CLL. In the trial reported by Brentjens et al., adult patients with refractory CLL with high disease burden were treated with the CD19-CD28z construct. The first four patients had no objective response and subsequently died of either progressive disease or infection. Authors concluded that the lack of clinical response was due to the omission of lympho depleting therapy; therefore, the subsequent cohort of four patients was pre-treated with cyclophosphamide. This group’s CARs underwent more robust expansion, and some of the patients achieved meaningful clinical responses that lasted 2-6 months [15].

Summary: This study supported the argument that lympho depleting chemotherapy plays an important role in creating the appropriate milieu for CAR proliferation without harmful effects on the host’s unmanipulated T cells, which may account for the difference in response rates between the two cohorts. Some of the theories are that lympho depleting chemotherapy depletes the host T cells including T regulatory cells which may interfere with CAR T cell proliferation.

Acute lymphoblastic leukemia (ALL)

Several early CAR studies were done in the setting of acute lymphoblastic leukemia. Most were small, single-institution phase 1 trials, and their treatment regimens varied greatly.

In a study of young patients reported by Maude et al., in 2014 [16], 30 children and young adults were treated with the CD19- 4-1BBz construct. Patients received different lympho depleting chemotherapies per physician choice, and CAR dosage ranged from 0.8-21 x 106 cells per kg body weight. Treatment was highly successful, with 90% of patients achieving complete remission and 73% minimal residual disease (MRD) negative status. Adverse side effects were common; cytokine release syndrome (CRS) occurred in all patients (with no CRS-related deaths reported), and 43% of patients suffered neurotoxicity. Overall survival (OS) at six month follow-up was 78.6%.

In another study reported by Davila et al. [17], sixteen adults with relapsed, refractory ALL were treated with the CD19-CD28z construct. Median patient age was 50 years, and all patients underwent lympho depleting pre-treatment with cyclophosphamide, 1.5-3.0 g/m2 , and CAR infusion at a dosage of 3 x 106 cells per kg body weight. Treatment was highly efficacious, with 88% of patients achieving complete remission and 75% MRD negative status. CRS occurred in all patients, and neurotoxicity affected 31% of patients. No deaths were reported.

In a third study reported by Lee et al. [18], 21 children were treated with the CD19-CD28z construct. CAR dosage ranged from 0.03-3.6 x 106 cells per kg body weight. Lympho depletion chemotherapy consisted of fludarabine 25 mg/m² per day on days –4, –3, and –2, and cyclophosphamide 900 mg/m² per day on day –2. Treatment resulted in a 60% complete remission rate, with 57% of patients achieving MRD negative status. CRS occurred in all patients, and neurotoxicity affected 29% of patients. In the 9.7-month follow-up period, event-free survival was 51.6%.

Summary: These three trials showed that CAR therapy could be safe and effective for patients spanning a wide age range and that complication such as CRS and neurotoxicity were generally treatable. Furthermore, these studies paved the way for multicenter phase 2 trials in pediatric and adult patients. Some of the ongoing clinical trials are listed in Table 1.

Table 1: Ongoing Clinical Trials of CAR T Cell Therapy in B Cell Lymphoma.

Clinical trial identifier

Phase

Title/Indication

Age (years)

CD19 CAR type

Gene transfer

Conditioning

Lead site

NCT02028455

I/II

CD19+ Leukemia

1-26

19-41BB

Gamma retrovirus

Not stated

Seattle

NCT02529813

I

Lymphoid Malignancies

1 - 80

 

 

Flu 25 or 30 mg/m2 Days -5 to -3

Cy250 or 300 or 500 mg/m2 Days -5 to -3

M.D. Anderson Cancer Center

NCT02030834

 

 

II

Refractory CD19+ Lymphomas

≥ 18

19-41BB

Lentivirus

Not stated

University of Pennsylvania

 

 

NCT02374333

I

Relapsed or Refractory CD19+ Leukemia and Lymphoma Previously Treated With Cell Therapy

1 - 24

19-41BB

Lentivirus

Not stated

University of Pennsylvania

NCT02640209

 

CART+ Ibrutinib In Patients With Relapsed Or Refractory CD19+ Chronic Lymphocytic Leukemia

 

≥ 18

19-41BB

Lentivirus

Not stated

University of Pennsylvania

NCT02050347

I

Relapsed CD19+ Malignancies Post-Allo HSCT

Any age

19-28

Gamma retrovirus

cyclophosphamide and fludarabine

Baylor College of Medicine

NCT01865617

I/II

CD19+ hematologic malignancies

≥ 18

19-41BB

Lentivirus

Not stated

Fred Hutchinson Cancer Research Center

NCT02146924

I

Acute Lymphoblastic Leukemia

≥ 18

19-28

Lentivirus

Not stated

City of Hope Medical Center

 

NCT02926833

I/II

DLBCL

≥ 18

19-28

Gamma retrovirus

cyclophosphamide and fludarabine

Multicenter

Abbreviations: CAR: Chimeric Antigen Receptor; DLBCL: Diffuse Large B Cell Lymphoma; r/r: Relapse Refractory; Cy: Cyclophosphamide; Flu: Fludarabine

Other lymphoid malignancies

Few studies have been completed in the setting of Diffuse Large B Cell Lymphoma (DLBCL). Kochenderfer et al. [19], presented a study of CAR therapy in patients with DLBCL (7 patients) and other B cell malignancies (8 patients). All received the planned dose of CARs (1-5 × 106 /kg), even though many were significantly lymphopenic. Of the seven patients with DLBCL, four achieved CR, two partial remissions (PR), and one stable disease. Of the patients with indolent lymphomas, all achieved either CR or PR. All but two patients had features of CRS or neurotoxicity. Authors concluded that CD19 CAR therapy in the setting of DLBCL and other B cell malignancies can be associated with significant response rates and a manageable toxicity profile. Porter et al. [20], also reported a series of 14 patients with CLL who received CAR therapy dosed at 0.14-11 × 108 cells per kg body weight. At follow-up (median duration 19 months, range 6-53 months), treatment had resulted in 28.5% CR and 28.5% PR;43% of patients did not achieve a meaningful response. All those who achieved CR were MRD negative, and authors concluded that there was potential for CAR therapy to eradicate CLL in patients who achieve MRD negative status.

There are also a few case series that document the effectiveness of CAR therapy in the treatment of multiple myeloma using CARs that target CD19, CD138, or B cell maturation antigen (BCMA) and the results have been variable ranging from partial to complete responses [21-24]. More carefully conducted phase 1 clinical trials are ongoing with CARs targeting BCMA.

One published report of a phase 1 trial of CD30 directed CAR therapy in 18 patients with relapse refractory Hodgkin lymphoma suggested that the therapy was well tolerated. Objective responses were noted in 39% of patients and the median progression free survival was 6 months. Toxicity profile was tolerable. Authors opined that the CD30 CAR therapy should be further studied in Hodgkin lymphoma [25].

ONGOING CLINICAL TRIALS

A detailed list of ongoing clinical trials is displayed in Table 1. The role for these early phase trials is to harmonize the treatment schedule of CAR therapy and to test the hypothesis that this innovative way of treating patients is feasible outside of highly specialized research laboratories. It remains to be seen whether CAR therapy could transition to the community setting.

SIDE EFFECTS OF THERAPY

The use of chimeric antigen receptor therapy has been associated with specific side effects whose pathophysiologies are often inherent to the mechanisms of CAR functioning. The most notable of these side effects are cytokine release syndrome (CRS), neurotoxicity/encephalopathy, and B cell aplasia.

Cytokine release syndrome

Cytokine release syndrome (CRS) is a reversible but potentially life-threatening condition thought to result from excessive pro inflammatory cytokine release and accompanying immune cell activation following rapid CAR activation and T cell expansion [16-19,26,27]. It is common in patients receiving CAR therapy (64-100% of patients in four major B-ALL and CLL trials were affected) and can present within days to weeks after CAR infusion [13,14,16-18,27]. Its presentation ranges in severity from mild, flu-like symptoms to high fevers, hypotension requiring volume resuscitation, capillary leak syndrome with hypoxia requiring supplemental oxygen or mechanical ventilation, and shock with multi-organ failure. Lab findings may be similar to those seen in hemophagocytic lympho histiocytosis

Table 2: Select Phase 1 Clinical Trials of CAR T Cell Therapy with at least 10 patients.

Author

Institution

Sample Size

Disease Type

CAR Type

Gene Transfer

Median Age (years)

Lymphodepleting Chemo

CAR T cell Dose

Response

Davila et al.

[17],

MSKCC

16

ALL

19-28

Gammaretrovirus

50

Cy

3 x 106/kg

88% achieved CR, 75% MRD negative. 44% bridges to allo-SCT

Maude et al.

[16],

UPENN

30

ALL

19-41BB

Lentivirus

14

Physician Choice

0.8 – 21 x 106/kg

90% achieved CR, 73% MRD negative. OS 78.6% at 6 months.

Lee et al.

[18],

NCI

21

ALL

19-28

Gammaretrovirus

 13

Flu 25mg/m2 Cy 900mg/m2

0.03 – 3.6 x 106/kg

68% achieved CR, 57% MRD negative. EFS 51.6% at 9.7 months

Porter et al. [20],

UPENN

14

CLL

19-41BB

Lentivirus

67

Flu/Cy 21%

Bendamustine 43%

Pen/Cy 36%

0.14 - 11x108

28.5% achieved CR, 28.5% PR and 43% did not respond

Abbreviations: ALL: Acute Lymphoblastic Leukemia; CLL: Chronic Lymphocytic Leukemia, CAR: Chimeric Antigen Receptor; CR: Complete Remission; SD: Stable Disease; ORR: Objective Response Rate; EFS: Event Free Survival; OS: Overall Survival; NA: Not Applicable; MRD: Minimal Residual Disease; Cy: Cyclophosphamide; Flu: Fludarabine; Pen: Pentostatin, MSKCC: Memorial Sloan Kettering Cancer Center; UPENN: University of Pennsylvania; NCI: National Cancer Institute.

 (HLH) and macrophage activation syndrome (MAS), including hyperferritinemia, hypofibrinogenemia, hypertriglyceridemia, and elevated C-reactive protein (CRP). Likewise, the cytokines characteristically elevated in CRS-IFN-gamma, TNF-alpha, soluble IL-2R alpha, IL-6, and IL-10-overlap with those implicated in HLH/MAS [13,27]. Daily monitoring of c- reactive protein (CRP) is recommended with CAR infusion to facilitate early identification of CRS; CRP in excess of 20 mg/dL during the time of CAR T cell expansion which occurs typically peaks between day 4-7 has been associated with high risk of severe CRS [17]. Offlabel treatment of CRS with tocilizumab, an IL-6 receptor blocker that spares CAR function, has shown clinical efficacy for symptom reversal [14,17]. Corticosteroids may cause CAR ablation with an associated increased risk of disease recurrence and should be reserved for refractory cases of CRS [13,28]. CRS occurrence, but not severity, is thought to reflect CAR efficacy, while symptom severity tends to correlate with tumor burden at the time of CAR infusion [13].

Neurotoxicity

CAR-related neurotoxicity varies in presentation and severity and tends to be progressive but reversible. Reported manifestations include dysphasia, confusion, global encephalopathy, akinetic mutism, and delirium, ranging to seizures, cerebral edema and coma. Seizure prophylaxis is recommended prior to CAR infusion [27], and patients with severe symptoms may require intubation for airway protection. Patients often must undergo imaging, EEG, and/or LP to rule out other causes of their symptoms. The mechanism of CAR-mediated neurotoxicity is poorly understood, but a theory of general inflammatory injury is favored over direct CAR T-mediated toxicity, as symptoms do not appear to correlate with either CAR presence in the cerebrospinal fluid or central nervous system(CNS)-lymphoma [17,27].

B cell aplasia

CD19-targeted CARs are designed to eliminate all CD19- bearing cells, including normal B cells. B cell aplasia is therefore termed an “on-target, off-tumor” toxicity and is considered a surrogate marker for CD19CAR efficacy [29,30]. Notably, hematopoietic stem cells, lacking CD19, are spared by CD19 CAR therapy and continue to manufacture B lymphocytes [31]. It follows that the duration of B cell aplasia corresponds with CAR persistence [32], the determinants of which are incompletely understood but thought to vary with tumor type, patient characteristics, conditioning regimen, and CAR construct [31]. In CD19-targeted CAR trials with high response rates in B-ALL, CARs have persisted for as little as one month to as long as two years [14]. Patients become severely hypogammaglobulinemic and can be managed with monthly infusions of intravenous immunoglobulins (IVIG) to decrease infection risk. Clinical monitoring of immunoglobulin levels has been used to identify IVIG requirement and as a proxy for detection of CAR depletion [32], which, if the patient is not in remission, may indicate tumor escape, poor CAR functioning, or a host immune response against the CAR.

FUTURE DIRECTIONS

CD19-directed CAR therapy has delivered on efficacy; response rates in the relapse setting are significantly higher than expected, especially considering that patients have more chemotherapy-resistant disease at the time of relapse. However, this efficacy has for the most part been restricted to very earlyphase clinical trials in a population that is subject to selection bias. It remains to be seen if these results are reproducible in ongoing phase 2 clinical trials and whether the described toxicities will make for a manageable safety profile. Once a tolerable safety profile is established in phase 2-3 multicenter trials, regulatory approvals will be sought to confirm the position of CAR therapy in the relapse setting. Given the range of prognoses for adult and pediatric patients with B-ALL, the future of CARs as first-line therapy is unclear and may vary depending on disease subtype. CARs will most likely emerge as an upfront treatment option for patients with poor prognoses for whom we do not currently have reliable therapies.

In the meantime, there are possible avenues for expanding the use of current CD19-targeted CARs, both in terms of retreating patients who have already received CAR therapy and enlarging the pool of eligible patients. For example, at the present time, active CNS lymphoma is a contraindication to most CAR therapies. There is concern that the phenomenon of neurotoxicity, which is poorly understood, may occur with greater severity in patients with known CNS lymphoma; thus, such patients have been excluded from clinical trials. However, as our understanding of CARs and the pathophysiology of their side effects improves, we may be able to develop specific protocols that would allow for treatment of patients with CNS involvement from systemic lymphoma, as well as those with primary CNS lymphoma.

Response to CD19CARs in different B cell malignancies has been variable, ranging from 80-90% in B-ALL to 45-76% in CLL [3,29,30]. The rapidity with which CD19CARs induce response and relative lack of definable antibodies to the CAR construct suggest that patients who tolerated therapy but only achieve durable response can be retreated with CARs. Determinants of response, however, remain unclear. Alteration of tumor antigen expression may explain why some patients fail to respond, become resistant to, or relapse after CAR therapy. As with other targeted therapies, tumor cells may escape CARs by down regulating or eliminating expression of the target antigen or through expansion of an antigen-negative sub clone. For example, antigen escape has been cited as a common cause of relapse in multiple trials of CD19-targeted CARs for B-ALL [31,33,34]. In an attempt to prevent this phenomenon, investigators have developed CARs that can recognize multiple antigens. In preclinical studies, so-called “bispecific” CARs were activated upon binding either CD19 or CD20, another pan-B cell marker, and showed robust antitumor response against both CD19-positive and CD19-negative lymphoma cell populations [34,35]. In the event that CD19-negative relapse does occur following CD19CAR therapy, follow-up treatment with CARs targeted to CD22, yet another pan B cell marker, have proven capable of inducing complete remission [31]. Similarly, in the setting of mature B cell malignancies, CARs targeted to the kappa light chain are showing promise in patients with CD19-negative relapse after CD19CAR therapy. An advantage to this approach is the persistence of lambda-B cells, such that patients are spared the B cell aplasia and severe hypogammaglobulinemia associated with CD19-targeted therapy [35].

Strategies for improving CAR specificity and decreasing toxicities have addressed multiple steps in CAR generation, including CAR gene delivery, mechanism of expression, structure, and co expression of other genes or CARs. Transfection of donor T cells with an inducible “suicide gene” enables rapid termination of graft versus host disease (GVHD) in patients receiving donor lymphocyte infusions after stem cell transplant for leukemia; in the context of CARs, a similar mechanism could provide an “off switch” to be employed in the case of severe adverse effects [2,10]. Alternatively, transfection of T cells with CAR mRNA, rather than DNA, allows for transient CAR expression and decreases the theoretical risk of mutagenesis associated with DNA integration into the genome [14,29,36]. Because the scFv portion of the CAR is typically of murine origin, patients may sometimes develop antimouse antibodies against the construct, with adverse impacts on CAR T functioning and persistence. Fully-humanized CARs have demonstrated efficacy, but self-tolerance to many target antigens, including CD19, makes selection of a suitable target difficult [37].

Preclinical and limited clinical data have suggested concurrent checkpoint inhibition may augment the efficacy of CAR therapy, both in hematologic malignancies and solid tumors [28,38-40]. Tumor cells are capable of evading immune system detection by exploiting the inhibitory T cell receptor programmed cell death protein-1 (PD-1), which normally functions as a feedback regulator on T cell response. By disrupting tumor cell PD-L1 engagement of PD-1 on T cells, PD-1 inhibitors like nivolumab and pembrolizumab prevent tumormediated T cell suppression. Combination PD-1 inhibitor/CAR therapy may therefore enhance CAR efficacy by averting CAR inactivation by tumor cells. Phase 1 and 2 trials combining CARs with pembrolizumab or an investigational PD-L1 monoclonal antibody, such as atezolizumab or durvalumab, are currently underway in patients with non-Hodgkin lymphomas as stated in Table 1 [39] Other philosophically similar approaches, such as CARs that have manipulated to express non functioning or modified PD-1 receptors, have also been associated with increased CAR activation [8]. Finally, combination CAR therapy with ibrutinib, a tyrosine kinase inhibitor, has been shown to decrease PD-1 expression by T cells and PD-L1 expression by CLL cells in patients with CLL [41].

Natural killer (NK) cells obtained from umbilical cord appear to have activity in acute myeloid leukemia (AML) and are being studies as a way of treating AML in the relapse setting [42-45]. There is evidence that NK cells can expand in the human body, secrete cytotoxic cytokines, and immunologically target acute leukemia cells. Meaningful responses have been described in mouse models [42]. Human studies are either planned or underway [42-45].

CARs have had limited success in the treatment of solid tumors. Difficulties in this setting include tumor architecture, which may prevent CARs from encountering many neoplastic cells, immunosuppressive tumor microenvironment, and selection of a target antigen that is widely expressed by the tumor and not by normal tissue [14,36]. Nevertheless, CAR trials for a range of solid tumors, including lung, breast, ovarian, colon, prostate, renal cell carcinoma, mesothelioma, glioma, and neuroblastoma, have been completed or are underway. The majority of these trials employ the standard CAR construct, targeted to a tumorspecific antigen such as CAIX (e.g., for renal cell carcinoma) [46]. However, the challenges solid tumors pose to traditional CARs have spurred CAR design modifications in the hopes of improving efficacy in this setting [36]. One example are the so-called “fourth-generation” CARs, whose activation additionally triggers pro-inflammatory cytokine release and innate immune system recruitment; this strategy is posited as a potential solution to solid tumor cells’ relative inaccessibility. Additionally, as innate immune cells may recognize and destroy tumor cells that have lost target antigen expression, these CARs may forestall tumor escape or relapse [47].

REFERENCES

1. Roeser JC, Leach SD, McAllister F. Emerging strategies for cancer immunoprevention. Oncogene. 2015; 34: 6029-6039.

2. Philip T, Guglielmi C, Hagenbeek A, Somers R, Van der Lelie H, Bron D, et al. Autologous bone marrow transplantation as compared with salvage chemotherapy in relapses of chemotherapy-sensitive non Hodgkin’s lymphoma. N Engl J Med. 1995; 333: 1540-1545. 

3. Hosing C, Saliba RM, McLaughlin P, Andersson B, Rodriguez MA, Fayad L, et al. Long-term results favor allogeneic over autologous hematopoietic stem cell transplantation in patients with refractory or recurrent indolent non-Hodgkin’s lymphoma. Ann Oncol. 2003; 14: 737-744.

4. Gokbuget N, Stanze D, Beck J, Diedrich H, Horst HA, Huttmann A, et al. Outcome of relapsed adult lymphoblastic leukemia depends on response to salvage chemotherapy, prognostic factors, and performance of stem cell transplantation. Blood. 2012; 120: 2032- 2041.

5. Thomas DA, Kantarjian H, Smith TL, Koller C, Cortes J, O’Brien S, et al. Primary refractory and relapsed adult acute lymphoblastic leukemia: characteristics, treatment results, and prognosis with salvage therapy. Cancer. 1999; 86: 1216-1230. 6. Essand M, Loskog AS. Genetically engineered T cells for the treatment of cancer. J Intern Med. 2013; 273: 166-181.

7. Marcus A, Eshhar Z. Allogeneic chimeric antigen receptor-modified cells for adoptive cell therapy of cancer. Expert Opin Biol Ther. 2014; 14: 947-954.

8. Goverman J, Gomez SM, Segesman KD, Hunkapiller T, Laug WE, Hood L. Chimeric immunoglobulin-T cell receptor proteins form functional receptors: implications for T cell receptor complex formation and activation. Cell. 1990; 60: 929-939.

9. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibodytype specificity. Proc Natl Acad Sci U S A. 1989; 86: 10024-10028.

10. Barrett DM, Singh N, Porter DL, Grupp SA, June CH. Chimeric antigen receptor therapy for cancer. Annu Rev Med. 2014; 65: 333-347.

11. Curran KJ, Pegram HJ, Brentjens RJ. Chimeric antigen receptors for T cell immunotherapy: current understanding and future directions. J Gene Med. 2012; 14: 405-415.

12. Davila ML, Bouhassira DC, Park JH, Curran KJ, Smith EL, Pegram HJ, et al. Chimeric antigen receptors for the adoptive T cell therapy of hematologic malignancies. Int J Hematol. 2014; 99: 361-371.

13. Dai H, Wang Y, Lu X, Han W. Chimeric Antigen Receptors Modified T-Cells for Cancer Therapy. J Natl Cancer Inst. 2016; 108. 14.Maude SL. Future directions in chimeric antigen receptor T cell therapy. Curr Opin Pediatr. 2017; 29: 27-33.

15. Brentjens RJ, Riviere I, Park JH, Davila ML, 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: 4817-4828.

16. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014; 371: 1507-1517.

17. 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.

18. 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: 517-528.

19. Kochenderfer JN, Dudley ME, Kassim SH, Somerville RP, Carpenter RO, Stetler-Stevenson M, et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol. 2015; 33: 540-549.

20. 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.

21. Ali SA, Shi V, Maric I, Wang M, Stroncek DF, Rose JJ, Brudno JN, et al. T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood. 2016; 128: 1688-1700.

22. 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: 1040-1047.

23. Jiang H, Zhang W, Shang P, Zhang H, Fu W, Ye F, et al. Transfection of chimeric anti-CD138 gene enhances natural killer cell activation and killing of multiple myeloma cells. Mol Oncol. 2014; 8: 297-310.

24. Gay F, D’Agostino M, Giaccone L, Genuardi M, Festuccia M, Boccadoro M, et al. Immuno-oncologic Approaches: CAR-T Cells and Checkpoint Inhibitors. Clinical Lymphoma, Myeloma & Leukemia 2017.

25. Wang CM, Wu ZQ, Wang Y, Guo YL, Dai HR, Wang XH, Li X, et al. Autologous T Cells Expressing CD30 Chimeric Antigen Receptors for Relapsed or Refractory Hodgkin Lymphoma: An Open-Label Phase I Trial. Clin Cancer Res. 2017; 23: 1156-1166.

26. Di Stasi A, Tey SK, Dotti G, Fujita Y, Kennedy-Nasser A, Martinez C, et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med. 2011; 365: 1673-1683.

27. Namuduri M, Brentjens RJ. Medical management of side effects related to CAR T cell therapy in hematologic malignancies. Expert Rev Hematol. 2016; 9: 511-513.

28. John LB, Devaud C, Duong CP, Yong CS, Beavis PA, Haynes NM, et al. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin Cancer Res. 2013; 19: 5636-5646.

29. Maus MV, Haas AR, Beatty GL, Albelda SM, Levine BL, Liu X, et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol Res. 2013; 1: 26-31.

30. Pegram HJ, Lee JC, Hayman EG, Imperato GH, Tedder TF, Sadelain M, et al. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood. 2012; 119: 4133-4141.

31. Khalil DN, Smith EL, Brentjens RJ, Wolchok JD. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol. 2016; 13: 273-290.

32. Tasian SK, Gardner RA. CD19-redirected chimeric antigen receptormodified T cells: a promising immunotherapy for children and adults with B-cell acute lymphoblastic leukemia (ALL). Ther Adv Hematol. 2015; 6: 228-241.

33. Jackson HJ, Brentjens RJ. Overcoming Antigen Escape with CAR T-cell Therapy. Cancer Discov. 2015; 5: 1238-1240.

34. Zah E, Lin MY, Silva-Benedict A, Jensen MC, Chen YY. T Cells Expressing CD19/CD20 Bispecific Chimeric Antigen Receptors Prevent Antigen Escape by Malignant B Cells. Cancer Immunol Res. 2016; 4: 498-508.

35. Ramos CA, Savoldo B, Torrano V, Ballard B, Zhang H, Dakhova O, et al. Clinical responses with T lymphocytes targeting malignancyassociated kappa light chains. J Clin Investigation. 2016; 126: 2588- 2596.

36. Zhang H, Ye ZL, Yuan ZG, Luo ZQ, Jin HJ, Qian QJ. New Strategies for the Treatment of Solid Tumors with CAR-T Cells. Int J Biol Sci. 2016; 12: 718-729.

37. Lanitis E, Poussin M, Hagemann IS, Coukos G, Sandaltzopoulos R,Scholler N, et al. Redirected antitumor activity of primary human lymphocytes transduced with a fully human anti-mesothelin chimeric receptor. Mol Ther. 2012; 20: 633-643. 

38. Cherkassky L, Morello A, Villena-Vargas J, Feng Y, Dimitrov DS, Jones DR, et al. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J Clin Investigation. 2016; 126: 3130-3144.

39. Chong EA, Melenhorst JJ, Lacey SF, Ambrose DE, Gonzalez V, Levine BL, et al. PD-1 blockade modulates chimeric antigen receptor (CAR)- modified T cells: refueling the CAR. Blood. 2017; 129: 1039-1041.

40. Larson RA, Sievers EL, Stadtmauer EA, Lowenberg B, Estey EH, Dombret H, et al. Final report of the efficacy and safety of gemtuzumab ozogamicin (Mylotarg) in patients with CD33-positive acute myeloid leukemia in first recurrence. Cancer. 2005, 104: 1442-1452.

41. Fraietta JA, Beckwith KA, Patel PR, Ruella M, Zheng Z, Barrett DM, et al. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood. 2016; 127: 1117-1127.

42. Cany J, van der Waart AB, Tordoir M, Franssen GM, Hangalapura BN, de Vries J, et al. Natural killer cells generated from cord blood hematopoietic progenitor cells efficiently target bone marrowresiding human leukemia cells in NOD/SCID/IL2Rg(null) mice. PloS one. 2013; 8

43. Langenkamp U, Siegler U, Jorger S, Diermayr S, Gratwohl A, Kalberer CP, et al. Human acute myeloid leukemia CD34+ CD38- stem cells are susceptible to allorecognition and lysis by single KIR-expressing natural killer cells. Haematologica. 2009; 94: 1590-1594.

44. Miller JS, Rooney CM, Curtsinger J, McElmurry R, McCullar V, Verneris MR, et al. Expansion and homing of adoptively transferred human natural killer cells in immunodeficient mice varies with product preparation and in vivo cytokine administration: implications for clinical therapy. Biol Blood Marrow Transplant. 2014; 20: 1252-1257.

45. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, Fautsch SK, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005; 105: 3051-3057.

46. Lamers CH, Klaver Y, Gratama JW, Sleijfer S, Debets R. Treatment of metastatic renal cell carcinoma (mRCC) with CAIX CAR-engineered T-cells-a completed study overview. Biochem Soc Trans. 2016; 44: 951-959.

47. Chmielewski M, Kopecky C, Hombach AA, Abken H. IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively Muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. Cancer Res. 2011; 71: 5697-5706

Wright FM, Oluwole OO (2017) Chimeric Antigen Receptor Therapy as a Means of Cellular Immunotherapy in B Cell Malignancies. J Cancer Biol Res 5(3): 1102.

Received : 15 Jul 2017
Accepted : 09 Aug 2017
Published : 18 Aug 2017
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X