Letter to the Editor Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jun 7, 2025; 31(21): 106530
Published online Jun 7, 2025. doi: 10.3748/wjg.v31.i21.106530
Targeting the NAD+/SIRT1 axis: A metabolic strategy to overcome oxaliplatin resistance in colorectal cancer
Md Sadique Hussain, Vikash Jakhmola, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehra Dun 248007, Uttarākhand, India
Kavita Goyal, Department of Biotechnology, Graphic Era (Deemed to Be University), Dehra Dun 248002, Uttarākhand, India
Arcot Rekha, Department of General Surgery, Dr. D.Y. Patil Medical College, Hospital and Research Centre, Pune 411018, Mahārāshtra, India
Ayesha Sultana, Department of Pharmaceutics, Sree Dattha Institute of Pharmacy, Hyderabad 501510, Telangāna, India
Haider Ali, Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, Tamil Nādu, India
Gaurav Gupta, Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
Gaurav Gupta, Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman 13306, United Arab Emirates
ORCID number: Md Sadique Hussain (0000-0002-3554-1750); Ayesha Sultana (0000-0003-3773-6604); Gaurav Gupta (0000-0001-7941-0229).
Co-corresponding authors: Md Sadique Hussain and Gaurav Gupta.
Author contributions: Hussain MS and Gupta G contribute equally to this study as co-corresponding authors; Hussain MS was responsible for conceptualization, data curation, writing – original draft; Jakhmola V was responsible for investigation, writing – original draft; Goyal K was responsible for formal analysis, investigation; Rekha A was responsible for data curation, writing – original draft; Sultana A was responsible for conceptualization, methodology, writing – original draft; Ali H was responsible for formal analysis, validation; Gupta G was responsible for conceptualization, supervision, writing – review & editing.
Conflict-of-interest statement: All the authors have no conflict of interest related to the manuscript.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Md Sadique Hussain, PhD, Assistant Professor, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Prem Nagar, Dehra Dun 248007, Uttarākhand, India. sadiquehussain007@gmail.com
Received: February 28, 2025
Revised: March 21, 2025
Accepted: April 16, 2025
Published online: June 7, 2025
Processing time: 98 Days and 7.5 Hours

Abstract

Oxaliplatin resistance remains a significant clinical challenge in colorectal cancer (CRC), highlighting the urgent need to identify novel molecular targets for therapeutic intervention. Recent findings by Niu et al have elucidated the role of the NAD+/SIRT1 axis in mediating oxaliplatin resistance through metabolic reprogramming. Their study demonstrated that oxaliplatin-induced DNA damage activates PARP, resulting in NAD+ depletion and subsequent downregulation of SIRT1. This reduction in SIRT1 levels enhances glycolysis, as evidenced by increased expression of PKM2 and LDHA, thereby conferring a metabolic advantage to resistant CRC cells. Conversely, restoration of SIRT1 expression reverses resistance, while pharmacological inhibition of glycolysis effectively sensitizes cells to oxaliplatin. These findings underscore the therapeutic potential of targeting the NAD+/SIRT1 pathway as a metabolic vulnerability in CRC. Future studies should investigate the clinical feasibility of combining SIRT1 agonists and glycolysis inhibitors with oxaliplatin to overcome drug resistance and improve patient outcomes.

Key Words: SIRT1; Glycolysis; Drug resistance; Colorectal cancer; Chemotherapy

Core Tip: Oxaliplatin resistance remains a major clinical challenge in colorectal cancer (CRC). This study underscores the critical role of the NAD+/SIRT1 axis in driving metabolic reprogramming that facilitates resistance. Oxaliplatin-induced DNA damage activates PARP, leading to NAD+ depletion and subsequent downregulation of SIRT1. This reduction in SIRT1 expression enhances glycolysis, marked by upregulation of PKM2 and LDHA. Notable, restoration of SIRT1 reverses resistance, while glycolysis inhibition sensitizes CRC cells to oxaliplatin. These findings suggest that targeting the NAD+/SIRT1 pathway, through SIRT1 agonists and glycolysis inhibitors, offers a promising metabolic strategy to overcome chemoresistance and improve therapeutic outcomes in CRC patients.



TO THE EDITOR

I am writing to express my strong interest in the recent study by Niu et al[1], which elucidates the role of the NAD+/SIRT1 axis in mediating oxaliplatin resistance in colorectal cancer (CRC). The findings presented in this study are particularly compelling as they offer novel insights into metabolic reprogramming as a mechanism of underlying chemoresistance. The interplay between DNA damage repair, NAD+ metabolism, and SIRT1 activity reveals a complex regulatory network that may serve as a promising therapeutic target to overcome oxaliplatin resistance in CRC patients. Given the growing clinical burden associated with oxaliplatin resistance, the therapeutic implications of targeting the NAD+/SIRT1 axis merits further discussion and investigation[1].

CRC remains a major oncological challenge, with oxaliplatin-based chemotherapy forming the cornerstone of treatment for advanced-stage disease. However, the emergence of resistance to oxaliplatin presents a formidable obstacle, significantly compromising treatment efficacy and reducing patient survival[2]. Although multiple mechanisms have been implicated in oxaliplatin resistance—including enhanced DNA repair capacity, increased activity of drug efflux pumps, and alterations in apoptotic signaling—the role of metabolic reprogramming has been relatively underexplored[3]. The study by Niu et al[1] provides compelling evidence that oxaliplatin-induced NAD+ depletion via PARP activation leads to the downregulation of SIRT1 expression, triggering a metabolic shift toward glycolysis. This adaptive response, characterized by elevated expression of PKM2 and LDHA, confers a survival advantage that enables CRC cells to evade oxaliplatin-induced cytotoxicity[1].

While the study elegantly links SIRT1 downregulation to glycolytic reprogramming, the precise molecular mechanisms through which SIRT1 modulates glycolysis warrant further elucidation. It remains to be determined whether SIRT1 directly regulates glycolytic enzyme function via post-translational modifications, such as deacetylation, or whether its effects are mediated through upstream signaling pathways. For instance, previous studies have indicated that SIRT1 may deacetylate and destabilize transcription factors like HIF-1α, thereby indirectly suppressing glycolytic gene expression[4,5]. Alternatively, SIRT1's role in activating AMPK and inhibiting mTOR signaling may intersect with key metabolic pathways critical for CRC cell survival under chemotherapeutic stress[6].

Importantly, the role of SIRT1 is context-dependent, influenced by both microenvironmental and genetic factors. While it may promote metabolic adaptation and chemoresistance in CRC, SIRT1 has also been shown to exhibit tumor-suppressive functions in other contexts. Therefore, a careful evaluation of potential off-target effects and CRC-specific molecular landscapes is essential to optimize the therapeutic targeting of the NAD+/SIRT1 axis.

Future investigations employing functional assays, such as chromatin immunoprecipitation to evaluate SIRT1’s direct binding to the promoters of glycolytic enzymes, and enzyme activity assays following SIRT1 modulation will be crucial to determine whether SIRT1’s influence on PKM2 and LDHA is direct or mediated through upstream regulators. Additionally, assessing the acetylation status of PKM2 and LDHA in oxaliplatin-resistant CRC cells could yield critical mechanistic insights.

Notably, prior studies have shown that SIRT1 agonists, such as resveratrol and SRT1720, exert anti-tumor effects in CRC models by modulating apoptotic pathways and cellular metabolism[7]. However, their specific impact on oxaliplatin resistance and glycolytic adaptation remains underexplored, highlighting the need for further investigation into SIRT1 modulation in this context.

The identification of SIRT1 as a key regulator of oxaliplatin resistance in this study aligns with growing evidence linking sirtuins to cancer metabolism and therapeutic resistance. SIRT1, an NAD+-dependent deacetylase, plays a pivotal role in maintaining genome stability, regulating apoptosis, and modulating cellular metabolism[8]. In the context of CRC, SIRT1 has been reported to exhibit dual functions, acting as either a tumor suppressor or an oncogene depending on the molecular and cellular context[9]. The current study suggests that NAD+ depletion, resulting from oxaliplatin-induced PARP activation, downregulates SIRT1 and enhances glycolytic activity, thereby promoting chemoresistance. Importantly, the finding that pharmacological activation of SIRT1 can restore oxaliplatin sensitivity underscores its potential as a viable therapeutic target.

An intriguing aspect of the study by Niu et al[1] is the elucidation of a link between DNA damage response (DDR) and metabolic adaptation. Oxaliplatin-induced DNA damage activates PARP, leading to NAD+ depletion and suppressing SIRT1 activity. This metabolic shift favors glycolysis over oxidative phosphorylation, mimicking the Warburg effect—a hallmark of cancer metabolism[10]. Notably, the study highlights how CRC cells exploit glycolysis reprogramming to survive chemotherapy-induced stress, thereby contributing to oxaliplatin resistance. The resensitization of resistant cells using glycolysis inhibitors, such as shikonin, underscores the therapeutic promise of targeting metabolic vulnerabilities in CRC[1].

While this study primarily addresses on oxaliplatin resistance, it raises an important question: Are SIRT1-mediated mechanisms also relevant to resistance against other platinum-based agents, such as cisplatin and carboplatin? Despite sharing the ability to induce DNA adducts, these agents differ in chemical structure, DNA-binding affinity, and associated repair pathways, all of which may influence how SIRT1 modulates cellular responses[11]. Cisplatin and carboplatin are extensively used in treating various malignancies—including ovarian, lung, and bladder cancers—where platinum resistance continues to pose a significant clinical challenge. Exploring the broader applicability of the NAD+/SIRT1 axis across different tumor types and platinum-based chemotherapeutics could inform the development of more effective combination strategies to overcome resistance.

Emerging evidence suggests that SIRT1 may similarly influence resistance mechanisms in other cancers. For instance, studies in ovarian and lung cancer models have implicated SIRT1 in regulating DDR, apoptosis, and metabolism—key processes involved in cisplatin sensitivity[12]. Therefore, future investigations should explore whether SIRT1 downregulation universally contributes to platinum resistance across tumor types. Integrating data from large clinical cohorts, such as The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus, could yield correlative insights into SIRT1 expression patterns and patient outcomes following cisplatin or carboplatin treatment. Additionally, functional studies in preclinical models utilizing various platinum agents would help determine whether the NAD+/SIRT1 axis constitutes a generalized metabolic vulnerability in platinum-resistant cancers.

The clinical relevance of these findings is underscored by data from TCGA, which indicates that lower SIRT1 expression is associated with poorer overall survival in CRC patients undergoing adjuvant chemotherapy[13]. This suggests that SIRT1 expression levels could serve as a prognostic biomarker to predict oxaliplatin response. Future studies should aim to validate these observations in larger patient cohorts and assess the therapeutic potential of SIRT1 agonists in clinical settings.

From a translational perspective, the study raises important questions regarding the potential for therapeutic intervention targeting the NAD+/SIRT1 axis. The use of SIRT1 activators, such as CAY10602, in combination with oxaliplatin represents a promising strategy to enhance chemosensitivity[14]. Moreover, considering the role of PARP activation in NAD+ depletion, the combination of PARP inhibitors with SIRT1 agonists may enhance therapeutic efficacy. Although PARP inhibitors have primarily been explored in BRCA-mutant cancers[15], their ability to influence NAD+ metabolism suggests a broader application in overcoming chemoresistance in CRC. Future preclinical and clinical studies are warranted to evaluate the feasibility and effectiveness of such combination approaches.

While the study by Niu et al[1] provides valuable mechanistic insights, several areas warrant further exploration to fully harness the therapeutic potential of targeting the NAD+/SIRT1 axis in CRC. First, the molecular determinants governing the differential response of CRC cells to SIRT1 modulation need to be elucidated. Given SIRT1’s dual role—as both a tumor suppressor and promoter depending on cellular and microenvironmental contexts—identifying predictive biomarkers (e.g., NAD+ levels, PARP activity, metabolic profiles) will be essential for patient stratification and optimizing therapeutic efficacy.

Second, the interplay between SIRT1 and other key metabolic regulators, such as AMPK and mTOR, should be systematically investigated. These pathways converge on cellular energy homeostasis and stress responses, and may synergistically influence glycolytic reprogramming and drug resistance. A comprehensive mapping of these interconnected signaling networks would provide a more holistic understanding of the metabolic adaptations that drive oxaliplatin resistance.

Importantly, while the current study emphasizes the benefits of pharmacological SIRT1 activation in restoring oxaliplatin sensitivity, the therapeutic potential of SIRT1 inhibitors in combination therapy remains underexplored. Emerging evidence suggests that SIRT1 inhibition may exacerbate DNA damage, oxidative stress, and apoptotic signaling—mechanisms that could enhance oxaliplatin cytotoxicity under certain tumor conditions[14]. It is therefore imperative to evaluate the efficacy of SIRT1 inhibitors in in vivo models, particularly orthotopic CRC models that more accurately replicate the tumor microenvironment, including stromal fibrosis, hypoxia, and immune infiltration.

Future preclinical studies should assess critical endpoints such as tumor growth inhibition, metastasis suppression, and systemic toxicity profiles in response to combination therapy with SIRT1 inhibitors and oxaliplatin. Mechanistic analyses should also investigate whether SIRT1 inhibition amplifies DNA damage markers (e.g., γH2AX), increases reactive oxygen species production, or enhances pro-apoptotic signaling in CRC cells. These insights would provide a rational basis for the dual targeting strategies and offer insight into the contextual benefits of either activating or inhibiting SIRT1. Collectively, such investigations will reinforce the translational relevance of NAD+/SIRT1 modulation and support the development of more effective, biomarker-driven combination therapies in oxaliplatin-resistant CRC.

Conclusion

The study by Niu et al[1] offers a compelling framework for understanding the metabolic underpinnings of oxaliplatin resistance in CRC, and positioning the NAD+/SIRT1 axis as a promising therapeutic target. Integrating metabolic interventions with conventional chemotherapy holds significant potential to enhance treatment efficacy and overcome drug resistance. Future research should prioritize the clinical validation of these findings and the development of combinatorial therapeutic strategies aimed to improving patient outcomes for CRC patients. In light of the urgent need for innovative approaches to address chemoresistance, targeting metabolic vulnerabilities emerges as a promising and clinically relevant avenue for advancing CRC therapy.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: India

Peer-review report’s classification

Scientific Quality: Grade A, Grade B, Grade B

Novelty: Grade A, Grade B, Grade B

Creativity or Innovation: Grade B, Grade B, Grade B

Scientific Significance: Grade A, Grade B, Grade B

P-Reviewer: Hasan N; Jin Y; Zhou Y S-Editor: Lin C L-Editor: A P-Editor: Yu HG

References
1.  Niu JW, Zhang GC, Ning W, Liu HB, Yang H, Li CF. Clinical effects of phospholipase D2 in attenuating acute pancreatitis. World J Gastroenterol. 2025;31:97239.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (1)]
2.  Capdevila J, Elez E, Peralta S, Macarulla T, Ramos FJ, Tabernero J. Oxaliplatin-based chemotherapy in the management of colorectal cancer. Expert Rev Anticancer Ther. 2008;8:1223-1236.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 30]  [Cited by in RCA: 29]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
3.  Chen G, Gong T, Wang Z, Wang Z, Lin X, Chen S, Sun C, Zhao W, Kong Y, Ai H, Yang H, Liu Y, Wu F, Kang J, Zhao S, Xiao X, Sun J, He A, Li Z. Correction to: Colorectal cancer organoid models uncover oxaliplatin-resistant mechanisms at single cell resolution. Cell Oncol (Dordr). 2022;45:1169-1170.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
4.  Joo HY, Yun M, Jeong J, Park ER, Shin HJ, Woo SR, Jung JK, Kim YM, Park JJ, Kim J, Lee KH. SIRT1 deacetylates and stabilizes hypoxia-inducible factor-1α (HIF-1α) via direct interactions during hypoxia. Biochem Biophys Res Commun. 2015;462:294-300.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 77]  [Cited by in RCA: 104]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
5.  Lim JH, Lee YM, Chun YS, Chen J, Kim JE, Park JW. Sirtuin 1 modulates cellular responses to hypoxia by deacetylating hypoxia-inducible factor 1alpha. Mol Cell. 2010;38:864-878.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 508]  [Cited by in RCA: 599]  [Article Influence: 39.9]  [Reference Citation Analysis (0)]
6.  Li J, Pan J, Wang L, Ji G, Dang Y. Colorectal Cancer: Pathogenesis and Targeted Therapy. MedComm (2020). 2025;6:e70127.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
7.  Sonnemann J, Kahl M, Siranjeevi PM, Blumrich A, Blümel L, Becker S, Wittig S, Winkler R, Krämer OH, Beck JF. Reverse chemomodulatory effects of the SIRT1 activators resveratrol and SRT1720 in Ewing's sarcoma cells: resveratrol suppresses and SRT1720 enhances etoposide- and vincristine-induced anticancer activity. J Cancer Res Clin Oncol. 2016;142:17-26.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 18]  [Cited by in RCA: 16]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
8.  Pande S, Raisuddin S. Molecular and cellular regulatory roles of sirtuin protein. Crit Rev Food Sci Nutr. 2023;63:9895-9913.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
9.  Dong W, Lu J, Li Y, Zeng J, Du X, Yu A, Zhao X, Chi F, Xi Z, Cao S. SIRT1: a novel regulator in colorectal cancer. Biomed Pharmacother. 2024;178:117176.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
10.  Barba I, Carrillo-Bosch L, Seoane J. Targeting the Warburg Effect in Cancer: Where Do We Stand? Int J Mol Sci. 2024;25.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 42]  [Cited by in RCA: 52]  [Article Influence: 52.0]  [Reference Citation Analysis (0)]
11.  Lokich J. What is the "best" platinum: cisplatin, carboplatin, or oxaliplatin? Cancer Invest. 2001;19:756-760.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 41]  [Cited by in RCA: 39]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
12.  Luna A, Aladjem MI, Kohn KW. SIRT1/PARP1 crosstalk: connecting DNA damage and metabolism. Genome Integr. 2013;4:6.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 67]  [Cited by in RCA: 79]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
13.  Guan J, Zhang ZY, Sun JH, Wang XP, Zhou ZQ, Qin L. LITAF inhibits colorectal cancer stemness and metastatic behavior by regulating FOXO1-mediated SIRT1 expression. Clin Exp Metastasis. 2023;40:309-320.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
14.  Yousafzai NA, Jin H, Ullah M, Wang X. Recent advances of SIRT1 and implications in chemotherapeutics resistance in cancer. Am J Cancer Res. 2021;11:5233-5248.  [PubMed]  [DOI]
15.  Pilié PG, Gay CM, Byers LA, O'Connor MJ, Yap TA. PARP Inhibitors: Extending Benefit Beyond BRCA-Mutant Cancers. Clin Cancer Res. 2019;25:3759-3771.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 194]  [Cited by in RCA: 254]  [Article Influence: 42.3]  [Reference Citation Analysis (0)]