The antileukemic effect of methylated indolequinone MAC681 involves degradation of PARP1 and immunogenic necroptosis

Increasing the tumor mutational burden by PARP1 inhibition and mitochondrial deregulation could add CML to the cancer types that could benefit from immunotherapy.

Published in Cancer and Cell & Molecular Biology

The antileukemic effect of methylated indolequinone MAC681 involves degradation of PARP1 and immunogenic necroptosis
Like

Share this post

Choose a social network to share with, or copy the URL to share elsewhere

This is a representation of how your post may appear on social media. The actual post will vary between social networks

Explore the Research

BioMed Central
BioMed Central BioMed Central

Antileukemic potential of methylated indolequinone MAC681 through immunogenic necroptosis and PARP1 degradation - Biomarker Research

Background Despite advancements in chronic myeloid leukemia (CML) therapy with tyrosine kinase inhibitors (TKIs), resistance and intolerance remain significant challenges. Leukemia stem cells (LSCs) and TKI-resistant cells rely on altered mitochondrial metabolism and oxidative phosphorylation. Targeting rewired energy metabolism and inducing non-apoptotic cell death, along with the release of damage-associated molecular patterns (DAMPs), can enhance therapeutic strategies and immunogenic therapies against CML and prevent the emergence of TKI-resistant cells and LSC persistence. Methods Transcriptomic analysis was conducted using datasets of CML patients’ stem cells and healthy cells. DNA damage was evaluated by fluorescent microscopy and flow cytometry. Cell death was assessed by trypan blue exclusion test, fluorescent microscopy, flow cytometry, colony formation assay, and in vivo Zebrafish xenografts. Energy metabolism was determined by measuring NAD+ and NADH levels, ATP production rate by Seahorse analyzer, and intracellular ATP content. Mitochondrial fitness was estimated by measurements of mitochondrial membrane potential, ROS, and calcium accumulation by flow cytometry, and morphology was visualized by TEM. Bioinformatic analysis, real-time qPCR, western blotting, chemical reaction prediction, and molecular docking were utilized to identify the drug target. The immunogenic potential was assessed by high mobility group box (HMGB)1 ELISA assay, luciferase-based extracellular ATP assay, ectopic calreticulin expression by flow cytometry, and validated by phagocytosis assay, and in vivo vaccination assay using syngeneic C57BL/6 mice. Results Transcriptomic analysis identified metabolic alterations and DNA repair deficiency signatures in CML patients. CML patients exhibited enrichment in immune system, DNA repair, and metabolic pathways. The gene signature associated with BRCA mutated tumors was enriched in CML datasets, suggesting a deficiency in double-strand break repair pathways. Additionally, poly(ADP-ribose) polymerase (PARP)1 was significantly upregulated in CML patients’ stem cells compared to healthy counterparts. Consistent with the CML patient DNA repair signature, treatment with the methylated indolequinone MAC681 induced DNA damage, mitochondrial dysfunction, calcium homeostasis disruption, metabolic catastrophe, and necroptotic-like cell death. In parallel, MAC681 led to PARP1 degradation that was prevented by 3-aminobenzamide. MAC681-treated myeloid leukemia cells released DAMPs and demonstrated the potential to generate an immunogenic vaccine in C57BL/6 mice. MAC681 and asciminib exhibited synergistic effects in killing both imatinib-sensitive and -resistant CML, opening new therapeutic opportunities. Conclusions Overall, increasing the tumor mutational burden by PARP1 degradation and mitochondrial deregulation makes CML suitable for immunotherapy.

Chronic myeloid leukemia (CML), a blood cancer characterized by the BCR-ABL fusion gene, presents ongoing challenges despite advancements in treatment. While tyrosine kinase inhibitors (TKIs) target the constitutively activated kinase, drug resistance and the persistence of leukemia stem cells (LSCs) remain significant obstacles. Approximately one in five patients develop resistance, with 22% becoming intolerant and requiring treatment discontinuation due to side effects (1, 2). Persistent LSCs exhibit altered mitochondrial metabolism, rendering them resistant to conventional therapies (3). In response to these challenges, we investigated a novel approach targeting DNA damage and metabolic rewiring. We demonstrated the role of non-apoptotic cell death in the context of improved immunogenic therapies against CML. Our research shows the importance of damage-associated molecular patterns (DAMPs) release and their implications for the immune response in CML treatment.

Our study began with an analysis of gene expression patterns in CML patients, revealing metabolic alterations and deficiencies in DNA repair pathways, including those associated with BRCA mutations. We found that PARP1, a key enzyme in DNA repair, was significantly elevated in CML stem cells compared to healthy cells (Figure panel 1). Building on this knowledge, we evaluated MAC681's potential to induce DNA damage (Figure panel 2) and disrupt mitochondrial function (Figure panel 3), leading to cell death. Besides rapid DNA damage induction, MAC681 also degraded PARP1, the essential DNA damage repair component in CML, further enhancing its efficacy against CML. Our in vitro and in vivo experiments confirmed MAC681's ability to reduce tumor growth.

We discovered that MAC681-induced degradation of PARP1 could be prevented by a known PARP1 inhibitor called 3-aminobenzamide (3-ABA). This finding is significant because 3-ABA is commonly used to inhibit PARP1, but our study suggests it may have unexpected chemical interactions with MAC681, potentially reducing its effectiveness. Therefore, caution should be taken when combining these types of compounds.

Furthermore, we found that MAC681-treated leukemia cells released DAMPs, which can enhance the immune response against cancer. Indeed, our experiments in mice showed that MAC681-treated cells could serve as an effective vaccine against CML, significantly reducing tumor growth (Figure panel 4). Additionally, we observed synergistic effects when combined with the novel TKI asciminib in both TKI-sensitive and -resistant models (Figure panel 5).

Overall, our study demonstrates the potential of targeting DNA repair, metabolic pathways, and inducing immunogenic cell death as a promising approach for treating CML. By disrupting key cellular processes and enhancing the immune response, compounds like MAC681 could offer new hope for patients with this challenging disease.

A mechanistic overview describing the effect of methylated indolequinone MAC681 in vitro and in vivo. The figure was created with BioRender.com.

References:

  1. Eide CA, O'Hare T. Chronic myeloid leukemia: advances in understanding disease biology and mechanisms of resistance to tyrosine kinase inhibitors. Curr Hematol Malig Rep. 2015;10(2):158-66.
  2. Geelen IGP, Thielen N, Janssen J, Hoogendoorn M, Roosma TJA, Willemsen SP, et al. Treatment outcome in a population-based, 'real-world' cohort of patients with chronic myeloid leukemia. Haematologica. 2017;102(11):1842-9.
  3. Kuntz EM, Baquero P, Michie AM, Dunn K, Tardito S, Holyoake TL, et al. Targeting mitochondrial oxidative phosphorylation eradicates therapy-resistant chronic myeloid leukemia stem cells. Nat Med. 2017;23(10):1234-40.

 

Please sign in or register for FREE

If you are a registered user on Research Communities by Springer Nature, please sign in

Follow the Topic

Cancer Biology
Life Sciences > Biological Sciences > Cancer Biology
Cancer Metabolism
Life Sciences > Biological Sciences > Cancer Biology > Cancer Metabolism
Cancer Therapy
Life Sciences > Biological Sciences > Cancer Biology > Cancer Therapy
Haematological Cancer
Life Sciences > Biological Sciences > Cancer Biology > Cancers > Haematological Cancer
DNA Damage Response
Life Sciences > Biological Sciences > Molecular Biology > DNA Damage and Repair > DNA Damage Response

Related Collections

With collections, you can get published faster and increase your visibility.

Multi-omics and Biomarkers

This collection aims to explore innovative methodologies and applications in integrating multi-omics data to identify and validate biomarkers.

Articles published in the collection have already gone through the systematic peer review process of the journal.

Publishing Model: Open Access

Deadline: Oct 31, 2025

Oncobiome

This collection of papers delves into the burgeoning field of oncobiome research, exploring the intricate relationship between cancer and the microbiome. The oncobiome encompasses the diverse microbial communities residing in and on the human body, which influence cancer development, progression, and treatment responses. By examining these interactions, our aim is to unravel the complex mechanisms through which the microbiome impacts oncogenesis and therapeutic outcomes. This compilation highlights cutting-edge research, offering insights into potential diagnostic markers and novel therapeutic strategies, thereby advancing our understanding of cancer biology and paving the way for innovative, microbiome-targeted cancer treatments.

Publishing Model: Open Access

Deadline: Ongoing