Fact-checked by Grok 2 weeks ago

MCL1

MCL1, or myeloid cell leukemia sequence 1, is an anti-apoptotic protein encoded by the MCL1 gene on 1q21.2 and belonging to the , where it promotes cell survival by sequestering pro-apoptotic effectors such as BAX and BAK at the mitochondrial outer membrane to inhibit . Discovered in 1993 during studies of differentiating human myeloblastic cells treated with phorbol esters, MCL1 was initially identified as an early inducible gene associated with myeloid cell survival and differentiation. The protein exists primarily as a 350-amino-acid isoform (MCL1L) with a short half-life of approximately 2–3 hours, featuring conserved BCL-2 homology (BH) domains (BH1–BH3, with a putative BH4), a C-terminal for mitochondrial localization, and an N-terminal sequence that facilitates rapid turnover via proteasomal degradation. Alternative splicing produces shorter variants like MCL1S and MCL1ES, which can exhibit pro-apoptotic functions by disrupting the full-length protein's activity. Beyond apoptosis regulation, maintains mitochondrial integrity, dynamics, and bioenergetics, while also influencing , progression (particularly the G2/M checkpoint), and . Its expression is tightly controlled at multiple levels: transcriptionally by factors such as and ; post-transcriptionally via microRNAs (e.g., miR-29, miR-125b) and long non-coding RNAs; and post-translationally through (e.g., by ERK or CDK1) and ubiquitination by E3 ligases like or β-TrCP, enabling rapid adaptation to cellular stress. is ubiquitously expressed at low levels in healthy tissues, with higher abundance in hematopoietic cells, neurons, and reproductive tissues, and it localizes mainly to mitochondria but also to the endoplasmic reticulum and nucleus. Dysregulated MCL1 overexpression is a hallmark of numerous cancers, including , , non-small cell , , and , where it drives tumor cell survival, proliferation, and resistance to chemotherapy and targeted therapies like inhibitors. In non-cancer contexts, MCL1 supports normal hematopoiesis, B-cell development, and tissue but has been implicated in pathologies such as , , and neurodegenerative diseases through impaired mitophagy. Therapeutically, selective MCL1 inhibitors such as S64315 (MIK665), AZD5991, and AMG 176 have shown preclinical efficacy in sensitizing cancer cells to ; however, as of 2025, clinical progress has been limited—AMG 176's phase I trial was terminated, AZD5991 demonstrated limited activity with cardiac safety concerns leading to a clinical hold, and S64315 is in ongoing combination trials—while newer inhibitors continue to enter early-phase studies for hematological malignancies, with persistent challenges including on-target due to MCL1's essential role in cardiomyocytes.

Overview

Discovery and Nomenclature

MCL1 was discovered in through studies on the ML-1 human cell line, where it was identified as an early-response during phorbol ester (phorbol 12-myristate 13-acetate)-induced along the / pathway. In these experiments, MCL1 mRNA levels increased rapidly within 1-3 hours of , preceding the appearance of differentiation markers by several days, suggesting its role in initial cellular responses to differentiation signals. The was isolated via differential screening of a constructed from mRNA of phorbol ester-treated ML-1 cells, using probes derived from subtracted cDNA to identify differentially expressed sequences. Upon sequencing, MCL1 was recognized as the first identified homolog of BCL2, a key regulator of apoptosis in lymphoid cells, based on significant sequence similarity in conserved domains. It was named "myeloid cell leukemia-1" (MCL1) to reflect its discovery in a myeloid leukemia cell line and its association with early stages of hematopoietic differentiation. This naming highlighted its potential relevance to myeloid malignancies, while establishing it as part of an emerging BCL2 gene family involved in cell survival. The nomenclature has evolved to include the full descriptive name "induced myeloid leukemia cell differentiation protein Mcl-1," emphasizing its induction during processes. The official symbol is MCL1 for the human and Mcl1 for the ortholog, as standardized by nomenclature committees. Key historical milestones include the cloning of full-length MCL1 cDNA from the induced ML-1 library, which enabled functional studies, and early experiments demonstrating that MCL1 overexpression in transfected fibroblasts prolonged cell survival under serum deprivation and other stress conditions, foreshadowing its anti-apoptotic role.

Gene Characteristics

The human MCL1 gene is located on the long arm of at band 1q21.2, with genomic coordinates spanning from 150,574,558 to 150,579,610 on the GRCh38 (complement strand). The gene covers approximately 5 and comprises four exons, encoding a protein-coding transcript as part of the . Its promoter region features binding sites for key transcription factors, including , which mediates induction in response to growth signals, and , which supports expression under inflammatory or stress conditions. Alternative splicing of the MCL1 pre-mRNA generates multiple isoforms, with the predominant long form (MCL1L, transcript variant 1; NM_021960.5) retaining all exons to produce a 350-amino-acid anti-apoptotic protein, while the short form (MCL1S, transcript variant 2; NM_182763.3) results from that introduces a frameshift, yielding a 271-amino-acid pro-apoptotic variant with a distinct . An additional extra-short isoform (MCL1ES; NM_001197320.2) has been identified, further diversifying potential regulatory functions at the RNA level. These isoforms arise primarily from at the 5' end and 3' , allowing context-dependent modulation of protein stability and activity. The MCL1 gene exhibits ubiquitous basal expression across human tissues, as evidenced by RNA-seq data from the GTEx consortium, but shows elevated levels in hematopoietic tissues such as peripheral blood mononuclear cells (TPM ~15.6) and lymph nodes (TPM ~13.2), as well as in the heart (TPM ~6.6) and liver (TPM ~6.1). This pattern underscores its critical role in rapidly proliferating or stress-exposed cell types. Expression is dynamically inducible by hematopoietic growth factors, including (GM-CSF), which rapidly upregulates MCL1 mRNA as an immediate-early response to promote progenitor survival. Similarly, cellular stresses such as , stress, and microtubule disruption trigger transcriptional activation to maintain viability. Evolutionarily, MCL1 demonstrates high sequence conservation across mammals, with over 230 orthologs identified in vertebrates, including the Mcl1 gene (NCBI Gene ID: 17210) on , where the protein shares approximately 90% amino acid identity with the human counterpart in the core functional domains. This conservation extends to non-mammalian vertebrates like , highlighting the ancient origin of MCL1's role in regulation, while variations in regulatory regions account for species-specific expression fine-tuning.

Protein Structure

Domains and Motifs

The MCL1 protein, in its predominant long isoform (MCL1L), comprises 350 amino acids and has a molecular weight of approximately 37 kDa, rendering it larger than most other BCL-2 family anti-apoptotic members owing to an extended N-terminal region spanning the first 170 residues. This N-terminal extension includes regulatory elements that distinguish MCL1 structurally from homologs like BCL-2 and BCL-XL. The core structure of MCL1 adopts a helical bundle typical of the BCL-2 family, consisting of eight α-helices, with a central hydrophobic α5 helix surrounded by amphipathic helices that enable membrane association. Central to MCL1's architecture are the BCL-2 homology (BH) domains that facilitate protein-protein interactions and heterodimerization. The BH3 domain, located at residues 209–223, forms part of the ligand-binding groove alongside contributions from other helices. The BH1 domain (residues 252–272) and BH2 domain (residues 304–319) contribute hydrophobic pockets essential for stability and binding specificity within this groove. A C-terminal transmembrane (TM) domain, approximately residues –350, anchors the protein to the outer mitochondrial , promoting its localization to organelles critical for cellular . These domains collectively define a BH3-binding groove wider than in other family members, influencing ligand affinity. MCL1 features distinctive N-terminal motifs that regulate its turnover and localization. -, -, serine-, and threonine-rich () sequences within the initial 170 residues confer a short protein that varies from approximately 30 minutes to 3 hours depending on and conditions, enabling rapid degradation via ubiquitin-independent pathways and allowing dynamic responses to cellular signals. An amphipathic in the core further supports membrane insertion beyond the TM domain, enhancing MCL1's association with lipid bilayers. Isoform variations arise from of the MCL1 gene. The short isoform, MCL1S (271 ), results from exon 2 skipping and lacks the BH1, BH2, and TM domains while retaining the BH3 region and partial . This structural truncation shifts MCL1S to cytosolic localization, where it mimics BH3-only pro-apoptotic proteins by antagonizing MCL1L function.

Comparison to BCL-2 Family

MCL1 belongs to the anti-apoptotic subgroup of the , which includes , , BCL-W, and A1/BFL-1, all of which inhibit by preventing mitochondrial outer membrane permeabilization (MOMP). These proteins share a common role in preserving mitochondrial integrity and promoting cell survival, particularly in response to stress signals. Structurally, MCL1 exhibits key similarities to other anti-apoptotics, including the conserved BH1, BH2, and BH3 domains that form a hydrophobic groove for binding pro-apoptotic BH3-only proteins, thereby regulating pore formation in the mitochondrial membrane. All family members, including MCL1, localize to the outer mitochondrial membrane via a C-terminal transmembrane (TM) domain, enabling their function at the site of MOMP. However, MCL1 possesses a flexible, unstructured N-terminal extension rich in PEST sequences, which is largely absent in and and facilitates its rapid proteasomal degradation. Additionally, MCL1's BH3-binding groove adopts a more open conformation with a shallower profile and distinct hot spots compared to the deeper, more constricted groove in , influencing its ligand selectivity—such as a preference for NOXA over other BH3-only proteins that bind more avidly to . Functionally, MCL1 diverges from its more stable relatives like , which have half-lives exceeding 24 hours, due to its short (<1 hour), allowing for dynamic of signals in proliferating cells such as lymphocytes. This instability enables MCL1 to provide transient anti-apoptotic protection tailored to acute stresses, contrasting with the constitutive support offered by in maintaining long-term cellular .

Biological Functions

Anti-Apoptotic Role

MCL1, an anti-apoptotic member of the , primarily exerts its function by sequestering pro-apoptotic BH3-only proteins such as BIM and , as well as the effector proteins BAK and BAX, thereby inhibiting mitochondrial outer membrane permeabilization (MOMP). This sequestration occurs through MCL1's hydrophobic groove, which binds the BH3 domain of these proteins, preventing their interaction with and activation of BAK and BAX on the outer mitochondrial membrane. By maintaining this binding, MCL1 blocks the oligomerization of BAK and BAX, which would otherwise form pores leading to the release of into the and subsequent activation of that execute . In healthy cells, MCL1 sustains mitochondrial integrity by continuously counteracting basal levels of pro-apoptotic signals, ensuring cell survival under normal conditions. Upon cellular stress, such as DNA damage or growth factor deprivation, BH3-only proteins are upregulated and displace BAK and BAX from MCL1, allowing these effectors to oligomerize and induce MOMP. This displacement triggers the intrinsic apoptotic pathway, highlighting MCL1's role as a sentinel that rapidly responds to survival cues to prevent unwarranted cell death. The structural basis for these interactions involves MCL1's BH3-binding cleft, which provides specificity for certain BH3-only proteins. MCL1 is particularly crucial for the survival of hematopoietic cells, including lymphocytes and plasma cells, where it promotes lineage commitment and prevents excessive during development and immune responses. For instance, in B-cell development, MCL1 supports the survival of pro-B and pre-B cells, while in plasma cells, it is indispensable for long-term production by inhibiting in these terminally differentiated cells. Genetic studies demonstrate that complete MCL1 in mice results in peri-implantation embryonic lethality around E3.5, primarily due to widespread in trophectoderm and developing hematopoietic tissues. Quantitatively, MCL1 protein levels show an inverse correlation with rates in hematopoietic cells; higher expression buffers against pro-apoptotic stimuli. Its short of approximately 20-30 minutes enables rapid degradation and replenishment, allowing quick adaptation to dynamic signals like IL-7, which stabilizes MCL1 in T-cells to enhance survival and memory formation during immune challenges. This turnover is mediated by ligases targeting MCL1's N-terminal domain, ensuring precise control over apoptotic thresholds.

Non-Apoptotic Roles

Beyond its canonical role in preventing , MCL1 exerts diverse non-apoptotic functions that influence cellular homeostasis and adaptation. In mitochondrial dynamics, MCL1 interacts with the optic atrophy 1 (OPA1) to stabilize its localization in the , thereby promoting mitochondrial fusion and maintaining cristae architecture essential for efficient . This interaction prevents excessive mitochondrial fragmentation under cellular stress, as MCL1 depletion disrupts OPA1 stability, leading to altered fusion-fission balance and compromised bioenergetic capacity. Conversely, in certain contexts, MCL1 also engages dynamin-related protein 1 (DRP1) to fine-tune events, ensuring mitochondrial network adaptability without triggering collapse. MCL1 further contributes to metabolic regulation by supporting fatty acid oxidation (FAO), particularly in metabolically demanding cells. In cancer cells, MCL1 binds acyl-CoA synthetase long-chain family member 1 (ACSL1) to facilitate long-chain and entry into mitochondrial β-oxidation, thereby sustaining production and . This function relies on MCL1's ability to preserve cristae integrity, as its matrix-localized isoform prevents structural disorganization that would impair efficiency. In immune cells, such as T lymphocytes, MCL1 is required for -induced metabolic reprogramming, where it promotes signaling to shift toward FAO-dependent persistence, enabling effector differentiation and formation. Additional non-apoptotic roles of MCL1 include suppression of and contributions to developmental processes. In fibroblasts exposed to , MCL1 inhibits through a specific loop domain spanning residues 188-207 without engaging apoptotic effectors. During early embryogenesis, MCL1 is indispensable for peri-implantation viability, supporting formation. Replacement with other members like can rescue this lethality. Similarly, in neutrophils, MCL1 ensures short-lived survival to maintain innate immune responses. Recent studies highlight that MCL1's non-apoptotic functions are essential for embryonic and postnatal , as substitution with purely anti-apoptotic members only partially rescues phenotypes. Tissue-specific effects highlight MCL1's contextual versatility. In cardiomyocytes, MCL1 preserves bioenergetic output by regulating mitochondrial calcium handling and cristae maintenance, preventing failure under hemodynamic stress. In neurons, the matrix isoform of MCL1 shields against —such as NMDA-induced damage—by enhancing respiratory chain function and inhibiting permeability transition pore opening, thereby promoting survival without direct of apoptotic effectors.

Regulation

Transcriptional Control

The MCL1 gene is transcriptionally regulated by a GC-rich promoter region that contains multiple binding sites for the transcription factors Sp1 and Sp3, which facilitate basal and induced expression in response to cellular signals. This promoter architecture allows MCL1 to respond dynamically to environmental cues, with Sp1/Sp3 sites enabling cooperative interactions with other regulatory elements. Several transcription factors bind directly to the MCL1 promoter to induce expression under specific conditions. binds to the MCL1 promoter, promoting upregulation during inflammatory responses, such as those triggered by IL-6 or IFN-α via the JAK/STAT pathway. , particularly the /p65 subunit, interacts with a site (GGGGTCTTCC) near the transcription start site (+35/+44 bp), activating transcription in response to pro-inflammatory or survival signals like through the MEK/ERK/ axis. Additionally, HIF-1α binds to a hypoxia-responsive element in the MCL1 promoter about 895 bp upstream, enhancing MCL1 expression during hypoxic stress to support cell survival. Induction of MCL1 transcription occurs through various signaling pathways activated by growth factors and stress. For instance, (EGF) and interleukin-6 (IL-6) stimulate expression via the MAPK/ERK pathway, which phosphorylates and activates downstream transcription factors that engage the promoter. (ER) stress similarly upregulates MCL1 via , helping cells adapt to proteotoxic conditions. Repression of MCL1 transcription is primarily driven by , which binds to intronic regulatory elements or the promoter in response to DNA damage, suppressing expression by up to 30-fold in a dose-dependent manner to promote . Post-transcriptional silencing also contributes to repression, with microRNAs such as miR-29 and miR-125b targeting the 3' (UTR) of MCL1 mRNA, reducing its stability and translation to lower protein levels; long non-coding RNAs can further modulate this by sponging miRNAs. In developmental contexts, MCL1 expression is elevated in proliferating cells but downregulated during terminal differentiation of myeloid lineages, ensuring appropriate control of cell survival during hematopoiesis.

Post-Translational Modifications

MCL1 protein stability and function are tightly regulated by multiple post-translational modifications (PTMs), which enable rapid responses to cellular signals due to its inherently short . These modifications primarily influence ubiquitination, degradation, and interactions with binding partners, allowing dynamic control over thresholds in proliferating or stressed cells. and ubiquitination represent the dominant PTMs, while and proteolytic cleavage provide additional layers of regulation. Phosphorylation of MCL1 occurs at specific residues within its PEST domain and surrounding regions, modulating its susceptibility to . GSK3β-mediated at Ser159 and Thr163 in the PEST region promotes MCL1 ubiquitination and subsequent proteasomal , a process activated during growth factor withdrawal or PI3K/AKT pathway inhibition. In contrast, ERK-mediated at Ser121, often alongside Thr163, stabilizes MCL1 during mitogenic signaling, enhancing its anti-apoptotic activity by inhibiting recognition. These opposing effects highlight 's role in balancing MCL1 levels based on survival cues. Ubiquitination targets MCL1 for proteasomal degradation, primarily at residues in the N-terminal domain, ensuring its rapid turnover. The E3 ubiquitin MULE (also known as ARF-BP1) initiates polyubiquitination of MCL1 under basal conditions, while TRIM17 acts as a neuronal-specific ligase that promotes degradation during induction. Deubiquitination by USP9X counteracts this process, stabilizing MCL1 and contributing to its overexpression in various cancers by prolonging its half-life. These enzymes collectively fine-tune MCL1 abundance in response to stress or oncogenic signals. Additional PTMs further diversify MCL1 regulation. at by the lysine acetyltransferase p300 reduces ubiquitination efficiency, thereby enhancing MCL1 stability and anti-apoptotic function, with this effect opposed by deacetylation from sirtuin 3. Caspase-3 and cleave MCL1 at Asp127 and Asp157, generating C-terminal fragments that lose anti-apoptotic activity and may promote by disrupting mitochondrial integrity or BH3-only protein sequestration. MCL1 exhibits a short basal half-life, typically 20-40 minutes to 1-2 hours depending on cell type and conditions, reflecting its dependence on continuous synthesis for maintenance, but this can extend to several hours under pro-survival conditions like ERK or USP9X activity. This kinetic profile is particularly critical in immune cells, where rapid MCL1 turnover facilitates quick adaptation to or signals, preventing unwarranted in dynamic environments.

Interactions

Binding Partners

MCL1 exhibits high-affinity binding to the pro-apoptotic effectors BAK and BAX, with a (Kd) of approximately 1 nM for BAK, while displaying moderate affinity for BID. These interactions occur through the BH3-binding groove of MCL1, forming stable complexes in cellular contexts. Among BH3-only proteins, MCL1 shows selectivity for NOXA, which binds with high potency and serves as an effective displacer of BAK from MCL1, alongside strong binding to BIM. In contrast, MCL1 has low affinity for and BAD, which preferentially interact with and . Beyond apoptotic regulators, MCL1 engages non-apoptotic partners such as OPA1 to support mitochondrial fusion and DRP1 indirectly through stabilization to influence fission dynamics. Additionally, MCL1 interacts with metabolic enzymes like ACSL1 in the oxidation pathway. Crystal structures reveal the NOXA BH3 helix docking into the MCL1 groove, as exemplified by the complex in PDB entry 2NLA, with cellular favoring 1:1 heterodimers for these interactions.

Inhibitor Development

Development of MCL1 inhibitors has focused on BH3 mimetics that disrupt the protein's anti-apoptotic function by targeting its BH3-binding groove, a shallow hydrophobic cleft that accommodates pro-apoptotic BH3 domains. Early efforts yielded non-selective compounds like , a natural BH3 mimetic derived from cottonseeds, which inhibits multiple members including MCL1, , and , with values in the micromolar range for MCL1. Despite its broad activity, gossypol demonstrated limited clinical success due to off-target effects and toxicity, paving the way for more selective agents. A milestone in selective inhibition came with S63845, developed by Servier and reported in 2016, which exhibits subnanomolar potency against MCL1 (Kd < 0.5 nM) and over 1,000-fold selectivity against other BCL-2 prosurvival proteins. This macrocyclic compound mimics the BH3 helix of NOXA, a natural MCL1-selective , and induces rapid in MCL1-dependent cancer cells, such as those from , , and models, both and . Preclinical studies highlighted its tolerability in mice, with efficacy in tumor xenografts at doses that spared normal tissues. Building on this, advanced small-molecule inhibitors have been tested in clinical stages, emphasizing hematologic malignancies where MCL1 overexpression drives resistance. AZD5991, from and disclosed in 2018, is a selective MCL1 with picomolar biochemical potency (Ki < 0.1 nM) that triggers release and in MCL1-reliant (AML) and myeloma cells. In a completed Phase I trial (as of 2024) for relapsed/ AML and myeloma, AZD5991 showed limited overall clinical activity and antitumor responses despite rapid tumor clearance in some responsive patients, constrained by on-target and other toxicities reflecting MCL1's essential role in platelet maturation and survival. Similarly, MIK665 (also known as S64315) from , a potent and selective MCL1 ( ~1.2 nM), was evaluated in completed Phase I trials (as of 2024), including combinations with or for AML and solid tumors; these showed synergistic induction in preclinical models but limited efficacy and managed hematologic toxicities in patients, with further development status unclear as of 2025. Design strategies for these inhibitors leverage structure-based approaches, informed by crystal structures of the MCL1 BH3 groove bound to NOXA or BIM peptides, to optimize hydrophobic interactions with key pockets (P2, P3, P4). Small molecules like S63845 and AZD5991 incorporate acylsulfonamide or cores to anchor deep , displacing bound effectors like BAK or BAX. Complementary peptide-based tactics include macrocyclic scaffolds and hydrocarbon-stapled alpha-helices that constrain BH3-like sequences from NOXA into rigid helical conformations, enhancing binding affinity (Kd ~1-10 nM) and cell permeability while selectively antagonizing MCL1 over or BCL-2. These strategies have yielded tool compounds for probing MCL1 dependency but face hurdles in translating to oral for clinical use. Key challenges in MCL1 inhibitor development include managing on-target toxicities, particularly transient and arising from MCL1's critical function in and survival, as observed in trials of S63845, AZD5991, and MIK665. Dosing regimens must balance efficacy with these effects, often requiring intermittent schedules to allow hematopoietic recovery. Additionally, compensatory upregulation of in response to MCL1 inhibition can confer resistance in tumor cells, necessitating combination strategies with inhibitors like navitoclax, though this exacerbates platelet toxicity. Ongoing efforts as of 2025 explore PROTACs and degraders to selectively deplete MCL1 while mitigating these liabilities, alongside novel antibody-drug conjugates incorporating MCL1 inhibitors as payloads.

Clinical Significance

Role in Cancer

MCL1 is frequently overexpressed in various malignancies, contributing to tumor cell survival by inhibiting . In hematologic cancers, amplification or gain of the MCL1 gene at 1q21 occurs in approximately 40% of cases, while high MCL1 expression is observed in nearly all newly diagnosed (AML) samples and is particularly prominent in chemotherapy-relapsed cases. In (DLBCL), about 10% of cell lines exhibit MCL1 mutations or copy number gains, with elevated levels correlating to higher tumor grades. Across solid tumors, 1q21 amplification drives MCL1 overexpression in up to 72% of breast cancers, 64% of lung adenocarcinomas, and around 12% of high-grade serous ovarian cancers. In tumorigenesis, MCL1 sustains survival following DNA damage induced by , thereby promoting chemoresistance across multiple cancer types. It plays an essential role in MYC-driven lymphomas by buffering , where MCL1 blockade effectively kills MYC-overexpressing cells, including those with mutations. High MCL1 expression serves as a negative prognostic marker; in non-APL AML, patients with elevated levels have a median overall survival of 4 months compared to 9 months in those with low expression, representing roughly a 55% reduction. According to DepMap data (DepMap Public 25Q3), MCL1 is a critical dependency in approximately 65% (776/1186) of profiled lines, underscoring its broad oncogenic relevance. Beyond hematologic malignancies, MCL1 supports in solid tumors by conferring resistance to anoikis, the triggered by from the , as demonstrated in and models where MCL1 depletion sensitizes cells to detachment-induced death and reduces metastatic potential. In HPV-driven , E6 oncoprotein-mediated upregulation of MCL1 enhances cell survival and contributes to oncogenesis.

Therapeutic Implications

MCL1 has emerged as a promising therapeutic target in cancer due to its frequent overexpression and role in promoting cell survival and therapy resistance. Clinical trials evaluating MCL1 inhibitors, such as AZD5991, have demonstrated limited efficacy as monotherapy in relapsed/refractory hematologic malignancies, including , with no objective responses observed in 12 patients and stable disease achieved in 41.7% of cases. However, these trials highlighted significant cardiac , including elevations in 83.1% of patients post-first cycle, leading to study termination and underscoring challenges in achieving durable responses without excessive . Combination strategies aim to enhance efficacy by addressing compensatory anti-apoptotic pathways. Preclinical studies show that pairing MCL1 inhibitors with , a inhibitor, synergistically induces in double-hit lymphomas by simultaneously disrupting MCL1 and BCL-2 dependencies, overcoming resistance seen in monotherapy. Early clinical data from trials like NCT03672695, evaluating S64315 (MIK665) with in and other malignancies, support improved antitumor activity, though full outcomes remain pending as of 2025. Biomarkers such as BH3 profiling identify MCL1-dependent tumors, predicting response to inhibitors, while the NOXA/BIM ratio influences sensitivity by modulating pro-apoptotic signaling and MCL1 neutralization. Beyond oncology, MCL1 modulation holds potential in non-cancer indications. In autoimmune diseases, selective inhibition of MCL1 in activated T-cells promotes , as demonstrated by sesamin's disruption of MCL1 function, offering a strategy to suppress pathogenic immune responses without broadly depleting regulatory T-cells. In neurodegeneration, enhancing MCL1 expression or activity protects neurons from mitochondrial dysfunction and , with CDK5 inhibition upregulating MCL1 to mitigate amyloid-beta-induced damage in Alzheimer's models. Future directions focus on next-generation agents to mitigate and improve specificity. PROTACs targeting MCL1 for ubiquitin-mediated , such as those based on S63845 warheads, show potent and selective activity in preclinical cancer models with reduced off-target effects compared to direct inhibitors. Completed trials, such as NCT02992483 for MIK665 in hematologic malignancies, along with other ongoing studies, provide updates as of 2025 on combination regimens and biomarker-guided patient selection to refine therapeutic applications.