The Case

A 22-year-old woman presented to an academic medical center with a four-week history of night sweats and tender progressive right cervical lymphadenopathy. A computed tomography (CT) scan of the neck revealed right cervical and supraclavicular lymphadenopathy (1.7 × 1.7 cm) with central necrosis. A soft tissue density (2.8 cm) in the anterior mediastinum was compressing the superior vena cava. A CT scan of the chest, abdomen, and pelvis revealed no enlarged lymph nodes and a normal-sized spleen. The complete blood count revealed a white blood cell count of 7.7 × 109/L; hemoglobin, 10.4 g/dL; hematocrit, 29 percent; platelets, 88 × 109/L; and absolute neutrophil count, 3.5 × 109/L. There were 20 percent blasts in the white cell differential count.

A cell suspension from a marrow aspirate was examined by flow cytometry, and the blast population expressed CD7, CD34, CD56, cytoplasmic CD3, human leukocyte antigen–DR, and partial CD15. The blast population was negative for CD1a, CD2, CD4, CD8, CD5 (dim), CD10, CD20, CD22, CD117, CD64, CD14, CD79a, and TdT. Chromosome analysis revealed a near triploid composite karyotype with 77 to 81 chromosomes. The molecular profile revealed no mutations in NOTCH1, FLT3 or IDH1, or IDH2. The spinal fluid did not contain leukemia cells. The lactate dehydrogenase was 327 U/L (normal range, 118-225 U/L). Her diagnosis was early T-cell precursor acute lymphoblastic leukemia (ETP-ALL). She was started on a standard four-drug induction regimen that included vincristine, dexamethasone, daunorubicin, and pegaspargase, including central nervous system prophylaxis.

Minimal residual disease (MRD) assessment was performed by multiparameter flow cytometry on day 29 of induction therapy and revealed a value greater than 0.1 percent. The patient continued on a pediatric consolidation regimen of augmented Berlin-Frankfurt-Munster (aBFM) protocols with the addition of nelarabine. Following completion of the consolidation course, MRD assessment was repeated and showed detectable disease (>0.1%). Given the high risk of relapse based on the end-of-consolidation MRD assessment, the patient was referred for allogeneic hematopoietic stem cell transplant (HSCT). As the patient was preparing for allogeneic HSCT, a marrow biopsy was performed as part of the pretransplant evaluation, and she was found to have 35 percent blasts in the aspirate specimen, consistent with relapsed T-cell ALL.

Current State of T-ALL Treatment/Challenges in Diagnosis and Management

T-lineage ALL (T-ALL) is curable for most children and adolescents and young adults (AYAs; defined as those aged 15-39 years), with contemporary frontline chemotherapy regimens.1,2  For children and AYAs, the pediatric-inspired, modified aBFM backbone including nelarabine has become the standard chemotherapy regimen in the United States.1  Despite the best initial chemotherapy, approximately 30 percent of children and adult patients will have MRD following induction and consolidation, a known risk factor for relapse,3  and approximately 15 percent of children and 25 percent of AYAs will have refractory disease or will relapse after an initial response.1,2  For adults 40 years and older, however, no comparably effective initial standard therapy exists. Because older adults cannot tolerate the use of high-intensity regimens inspired by pediatric studies, either dose-modified hyper-CVAD (cyclophosphamide, vincristine, doxorubicin, and dexamethasone),4  the regimen from the trial of the Medical Research Council in the UK and the Eastern Cooperative Oncology Group (E2993/UKALL12),5  or Cancer and Leukemia Group B protocols6,7  are commonly chosen. For adults with T-ALL treated on the E2993/UKALL12 study, the five-year incidence of relapse after initial response was 42 percent.5  Relapsed T-ALL is rarely cured in children or adults, with the overall survival rate for children being less than 30 percent, and for adults, less than 10 percent.5,8 

The major reason for the dismal prognosis after relapse is that current salvage therapies for T-ALL are ineffective for a majority of adults (60-70%) and too many children (30-40%), and no novel immunotherapy or targeted therapy has been established for patients with T-ALL. Unlike in B-lineage ALL (B-ALL), where immunotherapies targeted to CD19 or CD22 have significantly improved survival rates in relapsed disease, comparable therapies are not available or effective in T-ALL. Thus, for T-ALL, novel therapy is needed for 1) high-risk disease as defined by detectable MRD following induction and consolidation therapy, as described in the case report; 2) relapsed T-ALL, which has no effective salvage therapies; and 3) frontline treatment for older adults who cannot tolerate the intensity of pediatric regimens. This need has led to a burgeoning activity in the case of T-ALL to develop the same immunotherapies, including chimeric antigen receptor T-cell (CAR-T) platforms and monoclonal antibodies, as have been developed for B-ALL. This review will focus on these novel approaches and highlight immunotherapies currently in clinical trials. For the interested reader, several excellent reviews of T-ALL treatment and descriptions of genomic landscape have been published recently.9-12 

Monoclonal Antibodies

CD38 is a type II-transmembrane glycoprotein found on the surface of lymphocytes, including T and B lymphocytes, plasma cells, and natural killer cells. Its functions include regulation of intracellular calcium and signal transduction in immune cells. The laboratory of Dr. David Teachey and other labs have characterized CD38 expression in T-ALL to determine whether CD38 would be an effective target in this disease.13  The Teachey laboratory uncovered that blasts from pediatric patients with T-ALL have robust cell surface CD38 expression at the time of diagnosis, and that CD38 expression is maintained following chemotherapy. Further, there was low CD38 expression on normal lymphoid and myeloid cells and nonhematopoietic organs, suggesting minimal “off target” toxicity. The lab expanded the work to test daratumumab, a human immunoglobulin G1k monoclonal antibody that binds CD38 and is approved for myeloma therapy, and to determine its efficacy in patient-derived xenograft models of T-ALL. Daratumumab was highly effective in their model. In support of this approach are clinical case reports of compassionate use of daratumumab in relapsed T-ALL that have demonstrated prolonged responses.14  Collectively, the results support the likelihood that CD38 is a good therapeutic target for T-ALL and may be particularly useful in the clinical setting of detectable MRD following cytotoxic chemotherapy, as CD38 expression seems to be preserved following exposure to chemotherapy. To this end, daratumumab is being tested in combination with cytotoxic chemotherapy in an international multicenter phase I/II trial (NCT03384654) for children and AYA patients (ages 1-30 years) with relapsed or refractory B-cell and T-cell ALL and lymphoblastic lymphoma. Another monoclonal antibody targeting CD38, isatuximab, has been developed and was being tested as a monotherapy in a phase II trial (NCT02999633) for relapsed or refractory T-ALL or T-lymphoblastic lymphoma; however, the trial was closed permanently due to an unsatisfactory risk-benefit ratio. It is thought that the kinetics of response to anti-CD38 monotherapy are relatively slow, which is insufficient in an aggressive leukemia such as T-ALL. Therefore, the better strategy in this disease seems to be combining the antibody with cytotoxic chemotherapy. Thus, a second trial is ongoing in Canada and Europe for pediatric patients with either relapsed ALL or acute myeloid leukemia — a phase II study of the antibody in combination with cytotoxic chemotherapy (NCT0386044).

CAR-T Therapy

There are several hurdles to developing CAR-T therapy for the treatment of T-lineage leukemia. The complexity of harvesting T cells for the creation of the CAR-T construct results from the need to collect adequate numbers of normal T-cells from the patient without contamination of malignant T-cells. An alternative approach would be the use of genetically modified CAR-Ts from healthy allogeneic donors, an approach that may result in life-threatening graft-versus-host disease (GVHD) when the allogeneic cells are infused into immunocompromised patients.

Additionally, the targeting of lineage-restricted antigens on malignant T cells could result in two important adverse consequences: 1) fratricide, which in this scientific context refers to the nonspecific killing of the CAR-T construct; and 2) prolonged T-cell aplasia. Because the most widely expressed tumor antigen targets for malignant T cells are also expressed on normal T cells, the engineered T-cells can result in CAR-T fratricide during manufacturing and therefore limit ex vivo expansion and therapeutic potency of the autologous cell product. Once fratricide is resolved, there remains the issue of prolonged T-cell aplasia. CAR-T cytotoxicity against normal lymphocytes and their early precursors will suppress overall T-cell function and induce temporary or prolonged immunodeficiency, clinically similar to that observed following HSCT. There is no easy, clinically useful treatment to help with prolonged T-cell aplasia. Only time or potentially curative HSCT (as allogeneic HSCT terminates the activity of CAR-Ts) restores normal hematopoiesis and ultimately replenishes T-cell populations. In contrast, the prolonged B-cell aplasia from targeting CD19 in B-cell malignancies can be treated with intravenous immunoglobulin. Lastly, there is a risk of genetically modifying circulating malignant T-lymphoblasts, which could facilitate a treatment-resistant tumor clone.15 

Despite the technical and clinical challenges, several laboratories have developed CAR-T constructs, using strategies that address each of the technical and clinical hurdles described above, and early-phase clinical trials are underway (Table). Investigators at Baylor University have designed CAR-T constructs targeting CD5 and CD7 that eliminate fratricide, but these common T-lineage antigens will eliminate normal T-cell subsets and cause profound immunosuppression, so current trials are designed as a “bridge to transplant.”16,17  In the United Kingdom, scientists have used TRBC1 as a target, which is expressed in approximately 35 percent of T-ALL cases. This approach was designed to spare large subsets of normal T-cells to reduce T-cell aplasia.18  In an alternative approach, scientists at Washington University in St. Louis have designed an “off-the-shelf” CAR-T construct, UCART7, which is resistant to fratricide, exhibits no alloreactivity or GVHD potential, and expands and persists, and efficiently eliminates CD7+ T-ALL in vivo.19,20  The laboratory manipulations that were used to work around the obstacles are beyond the scope of this review, but are described in the references for the interested reader.

Selected Immunotherapy Trials for T-lineage Acute Lymphoblastic Leukemia

Selected Immunotherapy Trials for T-lineage Acute Lymphoblastic Leukemia
T-cell AntigenImmunotherapyTrial PhaseClinical Trial
CD5 CD5 CAR T Phase I NCT03081910/Baylor College of Medicine 
CD7 CD7 CAR T Phase I NCT03690011/Baylor College of Medicine 
CD7 UCART7 Phase I Not yet recruiting/Washington University 
TRBC1 TRBC1 CAR T Phase I NCT03590574/United Kingdom 
CD38 Daratumumab Phase I/II NCT03384654/Multiple international sites 
CD38 Isatuximab Phase II NCT0386044/Multiple international sites 
T-cell AntigenImmunotherapyTrial PhaseClinical Trial
CD5 CD5 CAR T Phase I NCT03081910/Baylor College of Medicine 
CD7 CD7 CAR T Phase I NCT03690011/Baylor College of Medicine 
CD7 UCART7 Phase I Not yet recruiting/Washington University 
TRBC1 TRBC1 CAR T Phase I NCT03590574/United Kingdom 
CD38 Daratumumab Phase I/II NCT03384654/Multiple international sites 
CD38 Isatuximab Phase II NCT0386044/Multiple international sites 

Conclusion

In summary, immunotherapy is a much-needed option for patients with resistant or relapsed T-ALL. The patient in the case report had few treatment options, especially given the initial use of nelarabine. She received mitoxantrone and cytarabine as salvage chemotherapy and did achieve a second remission. She is now more than 100 days post-HSCT and remains in remission. Her case is atypical, however, as most patients do not respond to salvage chemotherapy. The adult cooperative cancer groups, together with the children’s oncology groups, are working currently on the next series of clinical trials for T-ALL, some of which plan to incorporate daratumumab. CAR-T trials in progress may provide further progress toward an effective treatment for the significant fraction of children and adults who have relapsed or refractory T-ALL with current therapy.

1.
Dunsmore KP, Winter S, Devidas M, et al.
COG AALL0434: a randomized trial testing nelarabine in newly diagnosed T-cell malignancy.
J Clin Oncol.
2018;36:10500.
https://ascopubs.org/doi/abs/10.1200/JCO.2018.36.15_suppl.10500
2.
Stock W, Luger SM, Advani AS, et al.
A pediatric regiment for older adolescents and young adults with acute lymphoblastic leukemia: results of CALGB 10403.
Blood.
2019;134:1111.
https://www.ncbi.nlm.nih.gov/pubmed/31558558
3.
Schrappe M, Valsecchi MG, Bartram CR, et al.
Late MRD response determines relapse risk overall and in subsets of childhood T-cell ALL: results of the AIEOP-BFM-ALL 2000 study.
Blood.
2011;118:2077-2084.
https://pubmed.ncbi.nlm.nih.gov/21719599
4.
O'Brien S, Thomas DA, Ravandi F, et al.
Results of the hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone regimen in elderly patients with acute lymphocytic leukemia.
Cancer.
2008;113:2097-2101.
https://pubmed.ncbi.nlm.nih.gov/18720356
5.
Marks DI, Paietta EM, Moorman AV, et al.
T-cell acute lymphoblastic leukemia in adults: clinical features, immunophenotype, cytogenetics, and outcome from the large randomized prospective trial (UKALL XII/ECOG 2993).
Blood.
2009;114:5136-5145.
https://pubmed.ncbi.nlm.nih.gov/19828704
6.
Larson RA, Dodge RK, Burns CP, et al.
A five-drug remission induction regiment with intensive consolidation for adults with acute lymphoblastic leukemia: Cancer and Leukemia Group B Study 8811.
Blood.
1995;85:2025-2037.
https://pubmed.ncbi.nlm.nih.gov/7718875
7.
Larson RA, Dodge RK, Linker CA, et al.
A randomized control trial of filgrastim during remission induction and consolidation chemotherapy for adults with acute lymphoblastic leukemia: CALGB study 9111.
Blood.
1998;92:1556-1564.
https://pubmed.ncbi.nlm.nih.gov/9716583/
8.
Raetz EA, Borowitz MJ, Devidas M, et al.
Reinduction platform for children with first marrow relapse of acute lymphoblastic leukemia: a Children's Oncology Group Study.
J Clin Oncol.
2008;26:3971-3978.
https://pubmed.ncbi.nlm.nih.gov/18711187/
9.
Liu Y, Easton J, Shao Y, et al.
The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia.
Nat Genet.
2017;49:1211-1218.
https://pubmed.ncbi.nlm.nih.gov/28671688/
10.
Gianni F, Belver L, Ferrando A.
The genetics and mechanisms of T-cell acute lymphoblastic leukemia.
Cold Spring Harb Perspect Med.
2020;10a035246.
https://pubmed.ncbi.nlm.nih.gov/31570389/
11.
Hefazi M, Litzow MR.
Recent advances in the biology and treatment of T cell acute lymphoblastic leukemia.
Curr Hematol Malig Rep.
2018;13:265-274.
https://pubmed.ncbi.nlm.nih.gov/29948644/
12.
McMahon CM, Luger SM.
Relapsed T cell ALL: current approaches and new directions.
Curr Hematol Malig Rep.
2019;14:83-93.
https://pubmed.ncbi.nlm.nih.gov/30880359/
13.
Bride KL, Vincent TL, Im SY, et al.
Preclinical efficacy of daratumomab in T-cell acute lymphoblastic leukemia.
Blood.
2018;131:995-999.
https://pubmed.ncbi.nlm.nih.gov/29305553
14.
Ofran Y, Ringelstein-Harley S, Slouzkey I, et al.
Daratumomab for eradication of minimal residual disease in high-risk advanced relapse of T-cell/CD19/CD22-negative acute lymphoblastic leukemia.
Leukemia.
2020;34:293-295.
https://pubmed.ncbi.nlm.nih.gov/31435023
15.
Ruella M, Xu J, Barrett DM, et al.
Induction of resistance to chimeric antigen receptor T cell therapy by transduction of a single leukemic B cell.
Nat Med.
2018;24:1499-1503.
https://pubmed.ncbi.nlm.nih.gov/30275568/
16.
Mamonkin M, Rouce RH, Tashiro H, et al.
A T-cell directed chimeric antigen receptor for the selective treatment of T-cell malignancies.
Blood.
2015;126:983-992.
https://pubmed.ncbi.nlm.nih.gov/26056165
17.
Png YT, Vinanica N, Kamiya T, et al.
Blockade of CD7 expression in T cells for effective chimeric antigen receptor targeting of T-cell malignancies.
Blood Adv.
2017;1:2348-2360.
https://pubmed.ncbi.nlm.nih.gov/29296885
18.
Maciocia PM, Wawrzyniecka PA, Philip B, et al.
Targeting the T cell receptor beta-chain constant region for immunotherapy of T cell malignancies.
Nat Med.
2017;23:1416-1423.
https://pubmed.ncbi.nlm.nih.gov/29131157
19.
Gomes-Silva D, Srinivasan M, Sharma S, et al.
CD-7 edited T cells expressing a CD7-specific CAR for the therapy of T-cell malignancies.
Blood.
2017;130:285-296.
https://pubmed.ncbi.nlm.nih.gov/28539325/
20.
Cooper ML, Choi J, Staser K, et al.
An "off-the-shelf" fratricide resistant CAR-T for the treatment of T cell hematologic malignancies.
Leukemia.
2018;32:1970-1983.
https://pubmed.ncbi.nlm.nih.gov/29483708/

Competing Interests

Dr. O'Dwyer indicated no relevant conflicts of interest.