-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

4251 The AKR1C3-Activated Prodrug, Achm-025, Eradicates Disease in Preclinical Models of Aggressive T-Cell Acute Lymphoblastic Leukemia

Program: Oral and Poster Abstracts
Session: 614. Acute Lymphoblastic Leukemias: Therapies, Excluding Transplantation and Cellular Immunotherapies: Poster III
Hematology Disease Topics & Pathways:
Research, Translational Research, Non-Biological therapies, Chemotherapy, Combination therapy, drug development, Therapies
Monday, December 11, 2023, 6:00 PM-8:00 PM

Richard B. Lock, PhD1, Cara E Toscan1*, Hannah McCalmont1*, Kathryn Evans1*, Louise Doculara1*, Hansen J Kosasih1*, Andrew J Gifford1,2*, Amir Ashoorzadeh3*, Xiaojing Lin3*, Toby N Trahair1,4*, Charles E De Bock1*, Adam V Patterson3* and Jeff B Smaill3*

1Children’s Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
2Anatomical Pathology, NSW Health Pathology, Prince of Wales Hospital, Randwick, NSW, Australia
3Auckland Cancer Society Research Centre, Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland, New Zealand
4Kids Cancer Centre, Sydney Children’s Hospital, Randwick, NSW, Australia

Introduction: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy that is exceptionally difficult to cure after relapse. T-ALL expresses significantly higher levels of the enzyme aldo-keto reductase family 1 member C3 (AKR1C3) compared with B-cell ALL. To exploit this finding, we developed a novel prodrug, ACHM-025, which is selectively activated by AKR1C3 to form a potent cell-entrapped DNA alkylating agent. ACHM-025 was designed to improve drug specificity and minimize toxicity observed with currently used DNA alkylating agents, such as cyclophosphamide (CPM), a prodrug which is activated systemically via liver enzymes. We evaluated the in vivo efficacy and AKR1C3 selectivity of ACHM-025 against a panel of 25 pediatric T-ALL patient-derived xenografts (PDXs) alongside standard-of-care therapy.

Methods: AKR1C3 expression in T-ALL PDXs was determined by RNA-seq, immunoblotting and intracellular flow cytometry. For in vivo efficacy studies, PDXs were established as orthotopic models in immune-deficient NSG mice. Engraftment was assessed by enumerating the proportion of human versus mouse CD45+ cells (%huCD45+) in the peripheral blood. Treatment commenced when the %huCD45+ reached ≥1% (Day 0), and events were defined as %huCD45+ ≥25% or leukemia-related morbidity. Drug efficacy was assessed by mouse event-free survival (EFS) and stringent objective response measures.

For the single agent study, ACHM-025 (IP weekly, Days 0, 7, 14) was assessed against 25 T-ALL PDXs using a single mouse trial (SMT) format (one vehicle treated mouse, one drug treated mouse). For the consolidation therapy comparison, ACHM-025 (IP Days 0, 7) or CPM (IP Days 0, 7) combined with cytarabine (Ara-C; IP Days 0-4, 7-11) and 6-mercaptopurine (6MP; IP Days 0-4, 7-11) were assessed against a T-ALL PDX derived from a patient at relapse. For the relapsed/refractory (R/R) therapy study, ACHM-025 (IP Days 0, 7, 14) and nelarabine (IP Days 0-4, 14-18) were assessed against a T-ALL PDX derived from a patient at relapse.

Results: ACHM-025 dose-limiting toxicity in cynomolgus monkeys was neutropenia, where the pharmacokinetic equivalent dose in NSG mice was well tolerated. We first evaluated the in vivo efficacy of ACHM-025 as a single agent using the SMT format across an extended panel of 25 T-ALL PDXs, to address the impact of genetic heterogeneity in pediatric ALL on drug response. Remarkably, 7/25 PDXs treated with ACHM-025 did not relapse over 250 days after the last treatment and a total of 22/25 T-ALL PDXs scored an objective response. In comparison, vehicle treated EFS ranged from 3-26 days for the 25 T-ALL PDXs. Importantly, AKR1C3 expression (mRNA, protein or intracellular) provides a predictive biomarker of efficacy, as ACHM-025 was significantly more effective against T-ALL PDXs with high AKR1C3 expression versus those with low AKR1C3 expression (p<0.0001).

CPM is included in standard-of-care consolidation therapy for ALL in combination with Ara-C and 6MP. Comparing single agents, ACHM-025 was significantly more effective than CPM (T-C 25 vs. 57 days, p=0.0005, Table 1). The combination of ACHM-025, Ara-C and 6MP was significantly more effective than standard-of-care consolidation therapy (CPM, Ara-C and 6MP), more than doubling survival (T-C 36 vs. 75 days, p=0.0005, Table 1). Finally, we compared ACHM-025 with nelarabine, which is the only FDA approved agent for R/R T-ALL. Comparing single agents, ACHM-025 was substantially more effective than nelarabine (T-C 19 vs. 198 days, p=0.0005, Table 1). Importantly, relapsed disease remained sensitive to ACHM-025 re-treatment in vivo, with no evidence of acquired resistance. Remarkably, no mice treated with the ACHM-025/nelarabine combination relapsed over 250 days after the last treatment (Table 1).

Conclusions: ACHM-025 exerted profound in vivo efficacy against T-ALL PDXs and eradicated the disease in 7 aggressive T-ALL PDXs. ACHM-025 was significantly more effective than CPM both as a single agent and when used in combination with Ara-C/6MP. Notably, ACHM-025 in combination with nelarabine was curative when used to treat a chemoresistant T-ALL PDX in vivo. The in vivo efficacy of ACHM-025 directly correlated with AKR1C3 expression levels, providing a predictive biomarker for response. These data provide strong preclinical evidence highlighting the potential of ACHM-025 as a targeted and effective therapy for aggressive forms of T-ALL.

Disclosures: Ashoorzadeh: Lixin Pharmaceuticals: Patents & Royalties: Inventor on patent CN110809576B; Achilles Medical Co. Ltd.: Patents & Royalties: Inventor on patent US11661404B2; Health Innovation Ventures: Patents & Royalties: Inventor on patents DK2888227T3, EP2888227B1, US10202408B2, CA2886574C, US9873710B2, AU2013/306514B2, US9505791B2; Rain Oncology: Patents & Royalties: Inventor on patents CN107427515B, AU2015/358384B2, JP6769962B2, US10507210B2, CA2754808C, MX336332B, EP2406262B1, JP5925680B2, AU2010/290199B2, RU2568639C2, JP5793428B2, US9073916B2, US9101632B2, CN102574846B. Patterson: Achilles Medical Co. Ltd.: Patents & Royalties: Inventor of patent US11661404B2; Health Innovation Ventures: Patents & Royalties: Inventor of multiple patents (DK2888227T3, EP2888227B1, US10202408B2, CA2886574C, US9873710B2, AU2013/306514B2, US9505791B2); Rain Oncology: Current holder of stock options in a privately-held company, Patents & Royalties: Inventor of multiple patents (CN107427515B, AU2015/358384B2, JP6769962B2, US10507210B2, CA2754808C, MX336332B, EP2406262B1, JP5925680B2, AU2010/290199B2, RU2568639C2, JP5793428B2, US9073916B2, US9101632B2, CN102574846B); Lixin Pharmaceuticals: Patents & Royalties: Inventor on patent CN110809576B. Smaill: Achilles Medical Co. Ltd.: Patents & Royalties: Inventor on patent US11661404B2; Rain Oncology: Current holder of stock options in a privately-held company, Patents & Royalties: Inventor on multiple patents (CN107427515B, AU2015/358384B2, JP6769962B2, US10507210B2, CA2754808C, MX336332B, EP2406262B1, JP5925680B2, AU2010/290199B2, RU2568639C2, JP5793428B2, US9073916B2, US9101632B2, CN102574846B); Lixin Pharmaceuticals: Patents & Royalties: Inventor on patent CN110809576B; Health Innovation Ventures: Patents & Royalties: Inventor on multiple patents (DK2888227T3, EP2888227B1, US10202408B2, CA2886574C, US9873710B2, AU2013/306514B2, US9505791B2).

*signifies non-member of ASH