Physical Address
304 North Cardinal St.
Dorchester Center, MA 02124
Acute myeloid leukemia (AML) is a heterogeneous clonal disease of myeloid-stem and progenitor cells resulting from mutations, deletions, and epigenetic alterations in genes associated with cell differentiation, proliferation, and renewal. AML represents roughly 1% of all cancers; around 20,000 new cases and 11,000 deaths were projected to occur in the United States in 2021. AML encompasses a family of unique malignancies with over 5000 driver mutations across 76 genomic regions. Often, two or more of these driver mutations are present and genetically distinct clonal populations coexist. The stratification by these mutations, including FLT-ID3, NPM1, CEBPA, cKIT, DNMT3A, TP53, has formed a structure for therapeutic management.
The mainstay of treatment for young patients and those with favorable-to-intermediate prognoses remains a combination of cytarabine and anthracycline, while the optimal regimen for older patients has yet to be established. New therapies, including hypomethylating agents and receptor tyrosine kinase inhibitors, have shown promise in older patients with AML, and research is ongoing.
Although chemotherapeutic options for AML exist, T-cell–mediated allogeneic hematopoietic cell transplant (allo-HCT) remains the only potentially curative option, with 60% to 80% achieving complete remission. Even after successful engraftment of functional T-cells, however, AML cells retain the abilities to reduce expression of major histocompatibility complex (MHC) molecules, to change ligand expression to favor inhibition and quell activation, as well as to manipulate the tumor-immune microenvironment. These inherent capabilities allow leukemic cells to evade T-cell–mediated detection and elimination, leading to residual disease. Thus, even once remission is obtained, relapse exists as an ever-constant threat because of factors including chemoresistance and immune escape: relapse rates approach 40%, and 5-year overall survival is a dismal 20% to 30%.
In addition to identifying genetic mutations responsible for clonal evolution, research identifying markers for AML—including CD123, CLL-1, CD33, and NKG2D, among others—has energized the community toward the prospect of new and evolving therapies. Numerous investigators are working on checkpoint inhibitors PD-1 and CTLA-4, bispecific T-cell engagers, and chimeric antigen receptor (CAR) T-cells. Even with the current progress in targeted therapeutics, clinical outcomes remain unsatisfactory.
Significant advances have been made over the past decade in the field of adoptive T-cell therapy. Indeed, application of CD19 CAR-T-cell therapy has generated immense excitement in the fields of B-cell acute lymphoblastic leukemia and B-cell lymphoma and received U.S. Food and Drug Administration (FDA) approval in 2017. CAR T-cell technology is now investigated for implementation in other solid and liquid malignancies.
Regarding AML, advances in CAR-T-cell therapy have been limited by the paucity of tumor-specific antigens. Unlike B-cell malignancies, most myeloid leukemias share antigenic targets with healthy myeloid lineage cells. Various surface and signaling targets for myeloid leukemias have been proposed, and clinical trials are underway; however, the significant on-target, off-tumor effects on normal myeloid lineage as well as tissue cell populations lead to profound toxicity and remain a significant hurdle.
CARs are proteins created from the fusion of a variable extracellular antigen-binding domain (scFv) to the transmembrane and intracellular T-cell receptor (TCR) signaling domain, with addition of variable costimulatory moieties ( Fig. 14.1 ). CARs allow for antibody-mediated antigen targeting, thereby sidestepping limitations of MHC restriction and downregulation of natural costimulatory signal. This signal transmission promotes cytotoxic T-cell activation toward the target antigen, leading to T-cell–mediated tumor cell lysis. Thus, the ideal target for CAR-T-cells would be overexpressed by malignant cells and absent on healthy tissue.
The “first-generation” CARs expressed signal solely through the TCR CD3ζ domain without costimulation and demonstrated limited to no clinical effect. “Second- generation” constructs were later developed to include costimulatory domains CD28 or 4-1BB among others and demonstrated improved potency. “Third-generation” constructs contain a combination of costimulatory domains. The improvements in signal transduction and activation have led to higher-potency CAR-T-cells, and most current studies are focused on identifying antigen-binding domains to selectively unleash this potential power.
Though the first CAR trials targeting cancer began in the 1990s, the year 2013 marked one of the first phase I trials of CAR-T-cells in AML, targeting tumor-associated antigen Lewis Y. Since 2013, multiple targets have been proposed (including CLL-1, CD123, CD25, CD32, CD33, CD38, CD44, CD47, CD96, CD7, CD70, NKG2D, TIM3, and FLT3, among others). Many prospective targets are under investigation in Phase I/II/III clinical trials; presently there are 37 trials for the clinical application of adoptive cell therapy in the setting of AML ( Table 14.1 ).
Disease | Target | Eligibility | Identifier | Phase | Location |
---|---|---|---|---|---|
AML | Universal-CD123 | R/R AML | NCT03190278 | I |
|
CD123/CLL-1 | R/R AML | NCT03631576 | II/III | Fujian Medical University Union Hospital, China | |
CD123 | R/R AML | NCT04014881 | I | Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China | |
CD123 | R/R AML | NCT04265963 | I/II | 920th Hospital of Joint Logistics Support Force, China | |
CD123 | R/R AML | NCT03766126 | I | University of Pennsylvania, USA | |
CD123 | R/R AML | NCT04678336 | I | Children's Hospital of Philadelphia, USA | |
CD123 | R/R AML | NCT04272125 | I/II | Chongqing University Cancer Hospital, China | |
CD19 | R/R AML | NCT03896854 | I/II | The First Affiliated Hospital of Soochow University, China | |
CD19 | AML with t(8:21) | NCT04257175 | II/III | Chaim Sheba Medical Center, Israel | |
CD33 | R/R AML | NCT03971799 | I/II |
|
|
CD33 | R/R AML | NCT04835519 | I/II | Beijing Boren Hospital, China | |
CD38 | R/R AML | NCT04351022 | I/II | The First Affiliated Hospital of Soochow University, China | |
CD44v6 | AML or MM | NCT04097301 | I/II |
|
|
CD7 | R/R AML | NCT04762485 | I/II | The First Affiliated Hospital of Soochow University, China | |
CD7 | AML, T-ALL, NK cell lymphoma | NCT04033302 | I/II | Shenzhen Geno-immune Medical Institute, China | |
CD70 | R/R AML | NCT04662294 | I | The First Affiliated Hospital of Medical College of Zhejiang University, China | |
CLL-1/CD33 /CD123 | AML | NCT04010877 | I/II | Shenzhen Geno-immune Medical Institute, China | |
CLL-1 | Primary R/R AML | NCT04219163 | I | Texas Children's Hospital, USA | |
CLL-1 | R/R AML | NCT04884984 | I/II | The First Affiliated Hospital of Soochow University, China | |
CLL-1 | R/R AML | NCT04923919 | I | No.212 Daguan Road, China | |
CLL-1(KITE-222) | R/R AML and secondary AML | NCT04789408 | I |
|
|
FLT3 | R/R AML | NCT05023707 | I/II | The First Affiliated Hospital of Soochow University, China | |
FLT3 (TAA05) | R/R AML | NCT05017883 | N/A | Anhui Provincial Hospital, China | |
ILT3 | R/R AML (M4/M5) | NCT04803929 | I | Zhejiang Provincial People's Hospital, China | |
Universal-CAR γδT | R/R AML after transplantation | NCT04796441 | N/A | Hebei yanda Ludaopei Hospital, China | |
Universal-CD123 | R/R AML | NCT03190278 | I |
|
|
IL1RAP | AML at diagnosis and AML at relapse | NCT04169022 | N/A | CHU Besançon, France | |
B7-H3 (TAA06) | R/R AML | NCT04692948 | N/A | Anhui Provincial Hospital, China | |
|
CD123 | R/R AML, BPDCN, B-cell ALL, T-ALL | NCT04318678 | I | St. Jude Children's Hospital, USA |
Universal-CD123, TM123 | R/R AML, BPDCN, or B-cell ALL with CD123 expression | NCT04230265 | I |
|
|
CD123-CAR-CD28- CD3zeta-EGFR-T | R/R AML and Persistent/Recurrent BPDCN | NCT02159495 | I | City of Hope Medical Center, USA | |
|
CD33/CD38/CD56 /CD117 /CD123/CD34 /Muc1 | R/R AML or MDS | NCT03291444 | I | Zhujiang Hospital, Southern Medical University, China |
CD33 (PRGN-3006) | R/R AML or high-risk MDS | NCT03927261 | I | H Lee Moffitt Cancer Center and Research Institute, USA | |
NKG2D (CM-CS1 T) | AML or MDS, not in remission | NCT02203825 | I | Dana-Farber Cancer Institute, USA | |
NKG2D (CYAD-02) | R/R AML or MDS | NCT04167696 | I |
|
|
NKX101 CAR-NK | R/R AML and MDS | NCT04623944 | I |
|
|
|
CLL-1/CD33 | R/R AML and treatment-related AML, high-risk MDS, MPN, and CLL | NCT03795779 | I |
|
Interleukin-3 (IL-3) is a cytokine of the beta common family—including IL-5 and granulocyte macrophage colony-stimulating factor—which stimulates proliferation and differentiation of a range of hematopoietic cell types through interaction with the IL-3 receptor heterodimer (IL3-R). Notably, IL3-R is overexpressed by leukemic stem cells as well as by more differentiated blasts and thus has presented itself as a potential target. Importantly, however, IL3-R is also found on endothelial cells.
Multiple agents have been designed to target the IL3-R alpha chain (CD123). Notably, Tagraxofusp (formerly SL-401), a recombinant IL-3 linked to a diphtheria-toxin payload, has been approved for use in blastic plasmacytoid dendritic cell neoplasm (BPDCN), in which CD123 is ubiquitously overexpressed. In a study by Pemmaraju et al. for BPDCN, tagraxofusp led to a major clinical response in 90% of treatment-naïve patients, with complete response in 72%, ultimately leading to HCT for many patients; overall 24-month survival rate was 52%. In the same study, 67% of previously treated patients responded with median overall survival 8.5 months.
Determinants of response and resistance to CD123-targeted therapy represent an active area of research. Interestingly, Togami et al. explored tagraxofusp resistance and found that it is caused not by downregulation of expressed CD123 but by methylation and downregulation of a diphthamide pathway enzyme leading to resistance to the diphtheria-toxin payload. This methylation is reversible by the deoxyribonucleic acid (DNA)-methyltransferase inhibitor azacytidine. This study led to prolonged survival in vivo, and they have since initiated a phase 1 trial combining the two agents (NCT033113643).
Regarding CD123 targeting in AML, Tettamanti et al. and El Khawanky et al. demonstrated efficacy in xenograft models; however, more data is needed to assess safety and efficacy. El Khawanky et al. have also demonstrated success with combined azacytidine and CD123 targeted therapy: while CD123-CAR-T-cells did not entirely eliminate the leukemic cells, pretreatment of the recipients with azacytidine followed by CD123-CAR-T-cells led to long-term control, potentially via increased induction of CD123 by AML cells. Multiple clinical trials are investigating CD123-CAR therapy for AML (NCT04010877, NCT04318678, NCT04230265, NCT02159495, and NCT03291444).
Human C-type lectin molecule-1 (CLL-1) is a transmembrane glycoprotein present on 85% to 90% of myeloid lineage and leukemia stem cells. CLL-1 is also variably expressed by mature myeloid cells and absent on healthy hematopoietic stem cells (HSCs); this specificity contrasts with CD123 and CD33, which are present on HSCs. CLL-1-CAR-T-cell studies have yielded promising in vitro and in vivo results; multiple phase I and II clinical trials are ongoing (NCT04010877, NCT04219163, NCT04884984, NCT04923919, NCT04789408, and NCT03795779). Bu et al. have published encouraging results in pediatric AML with three patients achieving complete remission within 1 month of infusion. Zhang et al. reported a case with resultant 10-month response after CLL-1-CAR infusion and minor grade cytokine release syndrome (CRS). In another trial by Zhang et al., three of four pediatric patients treated with CLL-1-CAR infusion achieved complete remission while the other patient remained alive for 5 months. Clinical data so far suggest that CLL-1-CAR therapy has the potential to be safe and effective.
CD33, a transmembrane receptor molecule of the sialic acid binding immunoglobulin-like lectin family, is expressed by >90% of AML cells and to a lesser extent by healthy granulocytes. CD33 is potentially a promising target for AML.
Gemtuzumab-ozogamicin (GO) is a CD33-antibody-drug conjugate (ADC) bound to cytotoxic calicheamicin. GO initially garnered FDA approval in 2000 but was voluntarily withdrawn in 2010 after additional study data failed to demonstrate clinical benefit. After review of safety and efficacy, GO was again approved in 2017 after metaanalysis by Hill et al. indicated low-dose GO in combination with chemotherapy led to maintenance in 280 AML patients.
Regarding CD33-CAR-T-cell therapy, encouraging preclinical data has been published using the anti-CD33 scFv from GO. A phase I trial by Wang et al. following one patient treated with CD33-CAR-T-cells demonstrated marked decrease in AML blasts but profound myelosuppression 2 weeks post infusion. By 9 weeks, the patient had significant disease progression and ultimately died at 13 weeks. Tambaro et al. initiated a phase I trial (NCT03126864) to investigate third-generation CD33-CAR-T-cells with 11 patients with relapsed/refractory AML; ultimately 3 patients were infused, but no antileukemia responses were seen.
CD33 is also expressed by healthy myeloid cells, and studies have demonstrated marked myelosuppression with CD33-ADCs and CD33-CARs. Notably, CD33 is expressed on resident macrophages of the lung, liver, kidneys, and microglial cells, and hepatotoxicity is well documented. Multiple clinical trials are ongoing to further investigate the potential of targeted CD33 therapy (NCT04835519, NCT04010877 NCT03291444, NCT03927261, and NCT03795779).
Become a Clinical Tree membership for Full access and enjoy Unlimited articles
If you are a member. Log in here