Therapeutic inhibition of isocitrate dehydrogenase mutations in glioma and cholangiocarcinoma: new insights and promises—a narrative review
Introduction
Recent progress in cancer genetics have tried that hot-spot mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 may occur in lower-grade gliomas (LGGs), acute myeloid leukaemia (AML), intrahepatic cholangiocarcinoma (iCCA), chondrosarcoma, and thyroid carcinomas (1-8). Particularly, IDH2 mutations are also frequently evidenced in rare malignancies, such as angioimmunoblastic T-cell lymphoma and solid papillary carcinoma with reverse polarity (SPCRP) (9,10). Less frequently, these molecular alterations are reported in prostate tumors, paraganglioma, and melanoma (11-13). In particular, IDH2 mutation has been found recovered also in carotid body paragangliomas, a rare neuroendocrine tumor. This mutation may play a role in tumorigenesis of these tumors and could be used as a therapeutic target (14). IDH mutations occur frequently in chondrosarcoma, however, its prognostic role as well as therapeutic potential remain unclear (15,16). IDH mutation appears to play a central role in the establishment and maintenance of cancer stem cells (17).
The role of IDH mutation in cancer progression relies on its physiological function. As well established in literature evidence, IDH mutations, usually localised at the arginine residue (R132 for IDH1, R140, or R172 for IDH2), are somatic heterozygous and missense point mutations. The IDH mutated genes produce 51 the oncometabolite D-2-hydroxyglutarate (D-2HG) (18), thus inhibiting α-ketoglutarate (αKG)-dependent dioxygenases involved in the control of epigenetics and cellular growth and normal differentiation processes, in a manner that likely promotes leukemogenesis and tumorigenesis (19-21).
Therefore, intensive efforts have been conducted to identify small molecules with the ability to inhibit mutant IDH (mIDH) enzymes for the development of IDH-directed anti-cancer therapy (Table 1).
Table 1
Trial | Treatment | Phase | Setting | Status |
---|---|---|---|---|
NCT02746081 | BAY-1436032 | I | Advanced tumors, IDH1 mut | Active, not recruiting |
NCT02492737 | Vorasidenib (AG-881) | I/II | Advanced hematologic malignancies IDH1 and/or IDH2 mut | Completed |
NCT02481154 | Vorasidenib (AG-881) | I/II | Advanced tumors IDH1/2 mut (CCA, chondrosarcoma, glioma IDH172 mut) | Active, not recruiting |
NCT04164901 | Vorasidenib (AG-881) | III | Advanced malignancies IDH1r132 | Active, not recruiting |
NCT02074839 | Ivosidenib (AG-120) | I/II | Advanced hematologic malignancies with IDH1 mutation | Recruiting |
NCT02073994 | Ivosidenib | I/II | Advanced solid tumors (CCA, chondrosarcoma, glioma) | Active, not recruiting |
NCT03471260 | Ivosidenib + venetoclax + azacitidine | I/II | Advanced malignancies with IDH1 mutation | Active, not recruiting |
NCT03173248 | Ivosidenib + azacitidine | III | AML with IDH1 mutation | Recruiting |
NCT01915498 | Enasidenib (AG-221) | I/II | Advanced hematologic malignancies with IDH2 mutation | Active, not recruiting |
NCT02273739 | Enasidenib (AG-221) | I/II | Advanced solid tumors (glioma, iCCA, angioimmunoblastic T-cell lymphoma, chondrosarcoma IDH2 mut) | Completed |
NCT02381886 | IDH 305 | I/II | Advanced malignancies IDH1R132 | Active, not recruiting |
NCT04056910 | Ivosidenib + nivolumab | II | Advance tumors IDH1/2 mut | Recruiting |
NCT02428855 | Dasatinib | II | Advanced iCCA IDH1/2 mut | Completed |
NCT03878095 | Olaparib + ceralasertib | II | Advanced tumors IDH1/2 mut | Suspended |
NCT04521686 | LY3410738 | I | Advanced solid tumor IDH1 mut (glioma, CCA, chondrosarcoma, CCA, IDH2 mut) | Active, not recruiting |
IDH, isocitrate dehydrogenase; mut, mutant; CCA, cholangiocarcinoma; AML, acute myeloid leukemia; iCCA, intrahepatic cholangiocarcinoma.
In this regard, clinical results from randomised clinical trials showed the undoubtedly benefit gained in IDH-mutated LGG and cholangiocarcinoma with the use of anti-IDH targeted therapies (22-24). Unfortunately, nowadays, the use of these drugs is still not authorised in many countries due to local legislation.
Hence, in this review, we discuss the cancer-related role of IDH mutation and the results of mIDH-targeted therapies in the context of cholangiocarcinoma and gliomas highlighting promising developments, unanswered questions, and important future directions about this therapeutic approach. We present this article in accordance with the Narrative Review reporting checklist (available at https://cco.amegroups.com/article/view/10.21037/cco-24-17/rc).
Methods
We conducted a narrative review regarding the IDH1 and 2 mutations including the specific mutation and acquired resistance. In particular, we developed a research about the role of IDH1 and 2 mutation in cholangiocarcinoma and glioma tumors. We used the keywords: IDH1 mutation, IDH2 mutation, cholangiocarcinoma, glioma and resistance mechanisms to IDH1/2 mutations, filtered for last ten years and restricted to clinical trial, randomized trial and review and meta-analysis We used the PubMed searching system (Table 2).
Table 2
Items | Specification |
---|---|
Date of search | 23/July/2023 |
Database searched | PubMed |
Search terms used | IDH1 mutation, IDH2 mutation, resistance mechanisms to IDH1/2 mutations, glioma, cholangiocarcinoma |
Timeframe | 2009–2023 |
Inclusion criteria | All phase 2 and 3 trials, specific articles deal with IDH1 and 2 mutation and all selected articles in English are included |
Selection process | By all the authors, there is no need consensus because we find all in already published article |
IDH, isocitrate dehydrogenase.
An actionable mutation
Strength and limits
IDH1 resides in the cytosol, whereas IDH2 is located in the mitochondria. Despite their distinct cellular locations, both isozymes facilitate the reversible oxidative decarboxylation of isocitrate to αKG while simultaneously reducing NADP+ to NADPH. Through biochemical analyses, it has been observed that mIDH enzymes exhibit a neomorphic activity.
Specifically, they convert αKG into the oncometabolite D-2HG in a process that deviates from the norm by consuming NADPH rather than producing it, and concurrently generating NADP+ (25). The prognostic role of IDH1(R132) mutation is associated with reduced NADP+− dependent IDH activity in glioblastoma (25,26).
Physiologically, IDH is implicated in a range of cellular processes that go beyond metabolism to encompass epigenetic regulation, redox balance, and DNA repair mechanisms. One of the reaction products, αKG, serves as a cofactor for various enzymes collectively known as αKG-dependent dioxygenases. This group includes ten eleven translocation hydroxylases (TET), Jumonji c-domain containing lysine demethylase (KDMs), prolyl-hydroxylases domain-containing (PHD), factor inhibitor of HIF (FIH), and αKG-dependent-dioxygenase homologs (ALKBH) (27). TET family enzymes are involved in 5-methyl cytosine hydroxylation, while KDMs contribute to DNA and histone demethylation. These processes are pivotal for cell differentiation, influencing whether certain genes are expressed based on contextual transcriptional regulation (28).
The exact mechanism through which IDH mutations contribute to cancer pathogenesis remains uncertain, despite significant understanding of their biological impact. These effects are largely attributed to the structural resemblance between D-2HG and αKG, where the only distinction lies in the oxidation state of the carbon-2 position. This structural similarity leads to competitive inhibition, particularly among the extensive family of αKG-dependent dioxygenases, numbering over 70. Consequently, pathways utilizing αKG as a substrate are disrupted in IDH-mutant cancers. This disruption results in epigenetic dysregulation, marked by abnormal histone and DNA methylation, chromatin reorganization, hindered cellular differentiation, and other transformative effects (29-39).
Missense mutations, almost always heterozygous, in the IDH1 Arg132 codon cause a single amino acid substitution, most commonly to histidine (IDH1-R132H), but also to cysteine, serine, glycine, leucine, or isoleucine (7,12). Missense mutations in IDH2 codon for Arg140 or Arg172 (homologous to IDH1R132) occur predominantly as IDH2R140Q or IDH2R172K substitutions, although other amino acid changes occur (40,41). The common function of IDH1/2 active-site mutations is a neomorphic enzyme activity that catalyzes the conversion of αKG to D-2HG (18). Under physiological conditions, cellular D-2HG accumulation is limited due to the actions of the endogenous D-2HG dehydrogenase (D2HGDH), which catalyzes the conversion of D-2HG to αKG. However, the neomorphic activity of mIDH causes D-2HG to accumulate to supraphysiological levels within cells. Elevated D-2HG concentrations can be detected in the serum of patients with IDH-mutant AML and in IDH-mutant gliomas in patients (6,26,42-44). Elevated D-2HG levels in tumour tissues may provide a clinically useful biomarker for the non-invasive detection of IDH mutations due to the low background of D-2HG in normal tissue and almost invariable upregulation of D-2HG in the context of IDH active site mutations (45).
We can identify the most frequent IDH1 mutation with the help of immunohistochemistry using the anti-IDH antibody that recognises the R132H mutated protein. Less frequent IDH1 and IDH2 mutations can only be identified by DNA sequencing. IDH1 immunohistochemistry can be performed on fixed and paraffin embedded tissue samples. Concerning about cholangiocarcinoma, immunohistochemistry (IHC) positive mutations of IDH1 are not sufficient, but need next generation sequencing (NGS) or protein chain reaction (PCR) confirmation before starting a target therapy.
In situations where immunohistochemistry results are negative or inconclusive, especially in lower-grade tumors or suspected secondary glioblastomas, direct sequencing of IDH1 and IDH2 genes using PCR- or NGS-based methods should be carried out to screen for non-IDH1-R132H mutations. Non-invasive methods to detect the IDH mutation are being developed, including detection of the IDH1 mutation in plasma by digital PCR and magnetic resonance spectroscopy which allows the detection of abnormal accumulation of 2-hydroxyglutarate within the tumor, which could potentially help IDH mutation monitoring and therefore response to treatment (46,47). IDH1/2 RGQ PCR kits currently most used are Therascreen® (Redwood, CA, USA) and Entrogen® (Woodland Hills, CA, USA).
The epigenetic disruption caused by IDH1/2 mutant enzymes results in increased methylation of histones and DNA in a manner dependent on cell passage. This phenomenon is evident in the methylation profiles of various human malignancies, where tumors with IDH1/2 mutations exhibit a distinctive CpG island methylator phenotype. This phenotype is characterized by a significant level of DNA hypermethylation in CpG-rich regions (48). It has shown to both glioma and AML, epigenetic dysregulation caused by elevated D-2HG levels is reported to induce a DNA hypermethylation phenotype, which is clinically associated with increased methylation of patient tumor DNA and with the glioma-associated CpG island hypermethylated phenotype (GCIMP) (34,39,45-51). Several experimental and clinical data indicate that D-2HG-induced dysregulation of histone and DNA methylation suppress physiological cellular differentiation processes (35,52,53). Consequently, inhibition of cellular differentiation by D-2HG is thought to promote the pathological self-renewal of stem-like progenitor cells, which may create a cellular state that is permissive to malignant transformation. In addition to inhibiting αKG dependent dioxygenase enzymes, D-2HG has also been reported to activate, directly or indirectly, a number of enzymes and pathways (54-56). For example, D-2HG has been activated the prolyl hydroxylase egg-laying defective nine (EGLN) (55). Interestingly, this activation of EGLN appears to be a stereospecific effect of the D-2HG enantiomer, as (L)-enantiomer of 2-hydroxyglutarate (L2HG) is reported to inhibit EGLN activity (57). In preclinical models, expression of IDH1-R132H in myeloid cells determined splenomegaly, but also reduces bone marrow cellularity, and the authors observed the abundance of hematopoietic progenitor cells in a manner that corresponds with increased histone H3 methylation and CpG island hypermethylation (58). These data are confirmed by the observation that patient AML cells with IDH1 or IDH2 mutations show a common hypermethylated DNA phenotype and that transgenic expression of IDH2R140Q or IDH2R172K injurie the differentiation of 32D cells (a murine myeloid progenitor line) in cell culture (39). Collectively, evidence from AML patients and preclinical models strongly suggests that IDH1 and IDH2 mutations represent oncogenic drivers of AML and myelodysplastic syndrome (MDS) and that targeting IDH mutant neomorphic activity in this context may offer therapeutic benefit by promoting the differentiation of malignant myeloid cells (29,40). Like the results in hematopoietic model systems, expression of IDH1-R132H mutation in central nervous system (CNS) tissues has been highlighted to alter neurodevelopment and impair the differentiation of neural progenitor cells (59,60). Specifically, Sasaki et al. expressed IDH1-R132H under the control of the nestin or glial fibrillary acidic protein (GFAP) promoters in murine CNS cells and did not observe IDH1-R132H-dependent glioma formation (37). Instead, expression of mIDH1 resulted in perinatal lethality in all nestin and the majority (92%) of GFAP-promoter driven mIDH expression. The remaining percentage (about 8%) of GFAP-IDH1-R132H mice survived into adulthood and many of the surviving mice developed splenomegaly and liver tumors, likely due to leakiness of the GFAP promoter-regulated expression of Cre-recombinase in non-CNS tissues (39). A more recent study used a tamoxifen-inducible strategy to conditionally express IDH1-R132H in neural progenitor cells in 5–6 weeks old mice (61).
IDH-mutant inhibition
Given this biological reasoning, numerous research initiatives were conducted with the aim to discover therapies targeting IDH and explore their potential as anti-cancer medications. Preliminary investigations have demonstrated the in vitro effectiveness of IDH inhibitors. It has been demonstrated that in IDHmutant (IDHmut) glioma, there is an increasing of oxidative stress caused by reduction of NADPH production in glioblastoma cell line with IDH1(R132H) mutation. This seems to explain the increased overall survival (OS) in these patients, because the IDH1mut cells have less ability to survive to oxidative stress induced by ionization radiotherapy. These results may explain the longer survival of patients with IDH1-mutated tumors and that IDH1 inhibitor should not be used during radiotherapy (62).
The first IDH1 inhibitor, AGI-5198, was used by Popovici-Muller and colleagues in 2018. This molecule demonstrated a significant reduction (up to 90%) in 2-HG levels in a U87 glioblastoma xenograft mouse model. Subsequently, Rohle et al. found that AG-5198 not only inhibited 2-DHG but also induced the expression of differentiation markers, decreased cell proliferation and histone methylation within the same cell line. Despite these promising results, the suboptimal pharmacodynamic characteristics of AGI-5198 (i.e., rapid metabolism and clearance), have hindered its progression into clinical trials (63,64).
This molecule was followed by a series of inhibitory IDHs capable of binding to an allosteric site, stabilising the mutant enzyme in an open and inactive conformation, blocking the conformational change required for catalysis. Examples are specific inhibitors of mIDH1 (ivosidenib or AG-120; BAY-1436032) and mIDH2 (enasidenib; AG-221) as well as a dual inhibitor of mIDH1 and mIDH2 used both (vorasidenib; AG-881) in AML (65,66) (Table 3).
Table 3
Drug name | Target | Mechanism of action | Regulatory information-FDA | BBB penetration |
---|---|---|---|---|
Ivosidenib | IDH1 R132 C, H, G, S, L | Reversible, allosteric, competitive inhibitor | FDA approval for relapsed/refractory acute myeloid leukemia [2018] and for newly diagnosed [2019]. FDA approval for previously treated, locally advanced or metastatic cholangiocarcinoma with an IDH1 mutation [2021] | Unknown, 4.1% of penetrance in a rat model |
Enasidenib | IDH2 R140Q, R172K | Allosteric, non-competitive inhibitor | FDA approval for relapsed/refractory acute myeloid leukemia [2017] | No information |
Vorasidenib | Pan inhibitor, IDH1/2 | Allosteric, non-competitive inhibitor | FDA granted fast track designation to vorasidenib in IDH+ low-grade glioma [2023] | Penetrance in a rat model |
AG 5198 | IDH1 R132 C, H | Reversible, allosteric, competitive inhibitor to αKG | None | Penetrance in mouse glioma xenografts |
BAY 1436032 | IDH1 R132 C, H, G, S, L | Allosteric, non-competitive inhibitor | None | Penetrance (low): 0.06–0.38 brain to plasma ratio in a mouse model |
IDH, isocitrate dehydrogenase; FDA, Food and Drug Administration; BBB, blood-brain barrier; αKG, α-ketoglutarate.
Therefore, it is crucial to highlight the significance of IDH125, which possesses the ability to penetrate the blood-brain barrier (BBB), ensuring an optimal inhibition of D-2HG production. Additionally, IDH305 is a more soluble molecule with improved clearance kinetics and favourable inhibitory potency while IDH305, able to decrease the D-2HG production in IDH2 mutant cells to the same extent as IDH125, was tested in a clinical study (NCT02381886 trial). In this study patients with advanced-stage tumors carrying the IDH1 mutation were enrolled with promising early phase 1 safety results (67). DS-1001b, is another inhibitor currently under investigation in clinical trials (NCT03030066 and NCT04458272) involving patients with IDH1-mutant gliomas. It exhibits excellent BBB permeability and significantly decreases D-2HG production caused by IDH1-R132H and IDH1-R132C mutations. However, it shows limited effectiveness against IDH1-R132G, IDH1-R132L, and IDH1-R132S, and it does not inhibit IDH2 variants (68).
There are currently several phase I/II studies for new IDH inhibitors, such as the NCT02719574 study, which is evaluating the efficacy of the specific IDH1 mutant olutasidenib in patients with acute myeloid leukemia or MDS. Additionally, there is ongoing research on innovative “second-generation inhibitors”. Such as LY3410738 (NCT04521686 and NCT04603001) a molecule that not only forms a covalent bond with IDH but also robustly binds to IDH1 second-site mutations so conferring resistance to other inhibitors (69,70).
Crucially, it was observed by researchers that merely reducing D-2HG levels did not reliably indicate a clinical response. Many patients who did not respond to the therapy exhibited significant suppression of D-2HG. Instead, the researchers identified that mutations in genes associated with activating the RAS signalling pathway were more prevalent in non-responding patients (71) (refer to Table 1).
Resistance mechanisms
In spite of the optimistic findings outlined, there have been preliminary indications of acquired resistance mechanisms to these small inhibitors. This resistance leads to the advancement of the disease, accompanied by a rise in plasma 2-DHG concentration. Currently, ivosidenib and enasidenib have been approved for the treatment of IDH1- and IDH2-mutant AML, respectively. However, some patients harbouring IDH mutations show no response to monotherapy with mIDH inhibitors. Additionally, some patients experience relapse with increased circulating levels of 2-DHG and acquired resistance to IDH-targeted therapies. Among the initial instances of resistance, two patients with acute myeloid leukemia carrying the IDH2-R140Q mutation developed resistance to the IDH2 mutant inhibitor enasidenib. This resistance emerged following the occurrence of IDH2 second-site mutations, which can manifest in cis or trans (Q316E, I319M) in the wild-type allele. This collaboration with the gain-of-function mutation (R140Q) on the other allele induced resistance by disrupting the hydrogen bond between the IDH2 dimer and enasidenib or by hindering the binding of the IDH2 dimer to enasidenib. However, individually expressed, these second-site variants fail to generate D-2HG. Nonetheless, D-2HG production is restored upon co-expression with IDH2-R140Q, leading to the reinstatement of the self-renewal capacity of leukemic cells in vitro (72).
Mutations in the receptor tyrosine kinase (RTK) pathway may lead to primary resistance to ivosidenib (73). Conversely, the causes of acquired resistance can be multifactorial. In this context, the IDH1-R132C mutation was succeeded by an IDH1-S280F mutation, which is paralogous to residue I319 in IDH2. Other acquired second-site mutations in IDH1 include R199P, G131A, G289D, and H315D94. It is noteworthy that these mutations typically involve distinct subclones that emerge during treatment. In some cases, they may also be present at the treatment’s onset, highlighting the parallel expansion of multiple subclones with branching patterns and linear clonal evolution (74).
This resistance mechanism arises from the emergence of mutations in the opposite IDH isoform, which would still respond to D-2HG even during treatment with an IDH inhibitor. Furthermore, 2-HG induces hypermethylated Wnt inhibitory signals, with increased stemness. It follows that 2-HG resulting from IDH mutations enhances leukemia stemness, hindering cellular differentiation and leading to primary resistance to IDH inhibitors (75).
In summary, the development of new mutations in the opposing IDH isoform leads to resistance against mIDH1 or mIDH2 inhibitors, resulting in elevated D-2HG levels. This mechanism is under investigation in a clinical study involving the dual IDH1 and IDH2 inhibitor vorasidenib (NCT02492737). This molecule seems to be effective in the presence of known IDH1 second-site mutations due to its unique binding mode. However, it is still unclear how this drug prevents isoform switching. Another study with LY3410738 is ongoing. This molecule could be active in the context of known IDH1 mutations in the second site due to its unique mode of binding, which prevents isoform switching (76). D-2HG-restorative IDH second-site mutations still need to be investigated, as increases in D-2HG levels have been observed in IDH-independent mutants. It appears that some tumors have a predilection for D-2HG (77). Therefore, to improve the effectiveness of mIDH inhibitors and overcome these resistances, various therapeutic strategies are being studied. An alternative involves the use of targeted short hairpin RNAs, revealing a synthetic lethal relationship between BCL2 and IDH aberrations in AML cells. Enasidenib would reduce D-2HG levels, resulting in a reduction of cytochrome c oxidase activity. This process would lower the mitochondrial threshold for apoptosis activation following BCL-2 inhibition (78). Thus, in IDH-mutated hematological neoplasms, the association between the BCL-2 inhibitor venetoclax and ivosidenib (NCT03471260) is under study. In acute relapsed/refractory AML with IDH2 mutation (NCT04092179), a planned phase I/II was designed to investigate safety as well as effectiveness of enasidenib in combination with venetoclax (79).
In the phase III AGILE study (NCT03173248) and in other phase I/II studies, the combination of ivosidenib with hypomethylating agents such as azacitidine is under evaluation (80) (refer to Table 1).
IDH-mutant cholangiocarcinoma
Biliary tract cancers (BTCs) comprise a diverse group of aggressive malignancies that originate from various locations along the biliary tree, both inside and outside the liver. According to the updated anatomical classification, BTCs include iCCA, extrahepatic CCA (eCCA), which can be further categorized into perihilar (pCCA) and distal CCA (dCCA), as well as gallbladder cancer (GBC) and ampulla of Vater cancer (AVC) (81).
These tumors are often diagnosed at an advanced stage and typically do not respond well to chemotherapy, resulting in a grim outlook with a 5-year OS rate ranging from 7% to 20% (82). The underlying reasons for this aggressiveness have not been thoroughly explored. Currently, the only curative option for localized BTCs is surgical resection, but the recurrence rate remains high. Despite efforts to study the genomic characteristics of iCCA, there is still a lack of targeted therapies specifically designed for this subtype of liver cancer. It is crucial to gain a deeper understanding of the key driver events and evolutionary processes involved in ICC to develop more effective treatments (83). In recent years many studies have outlined a variety of molecular changes, including FGFR2, IDH1, HER2, MSI or BRAF, have the potential for targeted interventions.
The bile ductular and small duct subtypes of cholangiocarcinoma most commonly contain IDH1 mutations, specifically IDH1-R132C and IDH1-R132G, with a smaller percentage of cases having IDH2-R172 mutations. Missense mutation R132C is the most prevalent IDH1 mutation (44%) over the rest (84,85).
Numerous studies have evaluated the prognostic implications of these IDH mutations in cholangiocarcinoma, obtaining conflicting results regarding survival (86).
Several mouse models have investigated the effect of mIDH2 and demonstrated that IDH mutations alone do not generate hepatic or biliary lesions in vivo. However, the interaction of mIDH2 with the altered expression of transcription factors, oncogenes or tumor suppressors can generate tumor proliferation or aggressiveness in vivo (87). In 2014, Saha and colleagues conducted a study where they explored the control of the transcription factor SOX9, which is an early indicator of biliary cells within the liver. Their findings revealed that the regulation of SOX9 led to the downregulation of HNF4α, resulting in reduced differentiation of hepatocytes and an increase in cell proliferation in the liver (52).
In another in vivo experiment, which involved the introduction of IDH1-R132C, loss of p53 expression, and activation of Notch signaling within the liver, iCCA developed as early as 12 weeks after the injection of the Sleeping Beauty transposase expression vector. This model exhibited several characteristics associated with human iCCA, including the expression of CD19, the presence of collagen fibres, and active cell proliferation (88).
A potential drawback of previous experimental investigations into the roles of mIDH in the sustained growth of advanced solid malignancies is linked to the types of model systems employed. These models include xenografts, cell line or spheroid models, and engineered cancer models where ectopic mIDH1 expression doesn’t contribute to tumor initiation. These models may not fully capture important biological aspects of natural tumors. For this reason, in Wu et al.’s 2022 study, genetically engineered mouse models (GEMM) were presented to examine the functions of mIDH1 in the development of iCCA. Using a GEMM in which mIDH1 significantly promotes ICC development and related allograft models, a crucial pathway controlled by (R)-2HG was identified that coordinates epigenetic reprogramming and immune evasion. Inhibition of this pathway improved responsiveness to immunotherapy (89).
Interestingly, it’s worth noting that IDH1 mutations have been observed at a higher rate thus far, particularly in iCCAs that are not associated with hepatitis virus infection or Opisthorchis viverrini (90).
Just as they do in various other cancer types (several hematological and solid malignancies—such as glioma, iCCA, and acute myeloid leukemia), IDH mutations in cholangiocarcinoma disturb the typical differentiation patterns of hepatocytes and result in abnormal hypermethylation profiles triggered by D-2HG (91-93).
In particular, hypermethylation determines, most of the time, a downregulation of TET2 function, suggesting a phenotypic overlap in the same way as what is observed between IDH and TET2 mutations in AML; this data emerges from an analysis integrating whole-genome, transcriptomic and epigenomic data of 489 cholangiocarcinomas conducted by Jusakul et al. (94). Another study conducted in 2013 found that there are 5,758 CpG sites associated with 2,309 hypermethylated genes in IDH-mutant cholangiocarcinomas and that half of these genes are also hypermethylated in IDH mutant glioblastomas. Furthermore, 16 hypermethylated genes are underexpressed in both tumor types (95).
The presence of these common traits, rooted in similar genetic foundations across various tumors, indicates the existence of a shared mechanism in disease development. This, in turn, suggests the possibility of overlapping therapeutic strategies. Over the past few years, a considerable range of IDH inhibitors has been examined for their efficacy in IDH mutant cancers. Among these, ivosidenib (AG-120), first approved for the treatment of AML, stands out as the most advanced IDH inhibitor for CCA. A phase I clinical trial involving 73 individuals with advanced IDH1 mutant CCA led to the choice of 500 mg as the recommended dose of ivosidenib monotherapy. Furthermore, patients reported only objective, partial responses (PRs) or stable disease with significant reduction in plasma D-2HG levels compared to baseline levels. Two patients experienced the emergence of IDH mutations (specifically, IDH1-R132F and IDH2-R172V) during treatment, leading to resistance against the therapy (96).
More recently the phase III ClarIDHy study (NCT02989857) eligible patients with chemotherapy-refractory IDH1 mutated cholangiocarcinoma (70% with the IDH1-R132C mutation) received ivosidenib (n=124) or placebo (n=61). The treatment revealed a favorable safety profile showing median progression-free survival (mPFS) 2.7 months compared to 1.4 months with placebo and hazard ratio (HR) 0.37, 95% confidence interval (CI): 0.25–0.54; P<0.0001 while median OS (mOS) was unchanged between the two groups (97).
Saha and colleagues carried out additional preclinical investigations involving 17 BTC cell lines, screening them with 122 Federal Drug Administration (FDA)-approved drugs. Notably, the multi-target IDH tyrosine kinase inhibitor dasatinib demonstrated remarkable efficacy and specificity in targeting iCCA cells that carried IDH mutations. Other drugs, such as BCL-2 inhibitors and nicotinamide phosphoribosyltransferase (NAMPT) inhibitors active against other IDH-mutant tumors, were ineffective. Rous sarcoma oncogene (SRC) was identified as the main target of dasatinib in IDH-mutated cholangiocarcinomas thanks to the use of “gatekeeper” mutations that confer resistance to the drug in each of the inhibited kinases. Another phase 2 clinical study (NC T02428855) investigated the effect of dasatinib on 8 patients with mIDH CCA showing fair mPFS (8.7 weeks) and mOS (37.9 weeks) (98).
NCT03684811 it is instead a multicenter, phase Ib/II, open-label clinical study, investigating the mIDH inhibitor olutasidenib (FT-2102), in patients with relapsed or progressing solid tumors with IDH1-mutated. Between primary and secondary outcome measures are present objective response rate (ORR) to olutasidenib and recommended doses, PFS, time to progression, duration of response, OS, cerebrospinal drug level fluid and time to reach peak plasma concentration (99).
DNA damage response (DDR) alterations have also been described in CCA IDH-mutated due to their effect on aKG dioxygenases. Indeed, preclinical models have suggested the sensitivity of IDH mutant CCA cells towards PARP inhibitor (PARPi) could be enhanced by high levels of 2-HG. Therefore, studies of monotherapy with olaparib (NCT03212274) and of association olaparib (PARPi) with durvalumab (NCT03991832) were initiated.
Another phase 2 study is combining olaparib with ceralasertib (ATR inhibitor) in mIDH solid tumors (NCT038780950). However, the results obtained so far are not brilliant (100).
Moreover, the exploration of combining IDH inhibitors with chemotherapy or systemic immunotherapy presents an additional avenue currently undergoing assessment (refer to Table 1). Specifically, there is an ongoing phase I study focused on dose reduction, investigating the combination of ivosidenib and CisGem as a primary treatment for patients with metastatic disease (NCT04088188). On the flip side, there is an ongoing phase II clinical trial assessing the concurrent use of nivolumab and ivosidenib in advanced solid tumors carrying IDH1 mutations, which includes CCA (NCT04056910) (101).
IDH mutation in LGG
IDH1 is the most frequently mutated gene in low-grade gliomas (nearly 80% of cases), with the R132H mutation being the most frequently evidenced (102). Patients with grade 3 astrocytomas with IDHmut have a better prognosis than IDH wild type (IDHwt) astrocytomas (mOS 51 vs. 22 months) (102).
Moreover, IDH mutations provide the G-CIMP hypermethylated state and a proneural gene-expression profile (103,104). In this regard, although the immunosuppressive nature of gliomas has been well documented (105), the exact roles of mIDH and D-2HG production in immunosuppression remain unclear and under evaluation. Precisely, the overall amount of tumor-infiltrating lymphocytes is lower in IDH-mutant versus IDH-wild-type gliomas (105). Furthermore, subsequent analyses of particular immune cell subsets revealed the presence of a global reductions of microglia, macrophages, dendritic cells, B cells and T lymphocytes (106).
Additionally, it is well known that the selective permeability of the BBB represents a selective barrier to the delivery of systemic therapies in the CNS (107).
In this regard, the expression of ATP-binding cassette transporters in tumour cells often results in the efflux of drugs that do penetrate the BBB, that even further can prevent drug delivery.
These difficulties must be taken into account during drug development to achieve effective glioma targeting. In this regard, patients with glioma were included in a phase I multi-centric basket trial focused on the safety of ivosidenib used in patients with IDH1-mutant solid tumors (NCT02073994). In general, the molecule was well tolerated, with 13 (19.7%) of 66 patients with advanced-stage gliomas having grade ≥3 adverse events (AEs), and they observed only two AEs related to the treatment. Regarding efficacy results, among the 35 patients with non-enhancing lesions, the ORR was 2.9%, with one PR. In this group the authors achieved a stable disease in 85.7% of patients (30) compared with 45.2% (14) of those with enhancing gliomas. The authors achieved a mPFS of 13.6 months for patients with non-enhancing gliomas and 1.4 months for those with enhancing lesions.
Twenty-two of 33 patients experienced a reduced tumor volume and 9 (33.3%) of 27 evaluable patients with non-enhancing and enhancing lesions, respectively; patients with non-enhancing lesions, the authors demonstrated that the estimated tumor growth rate in 6 months decreased from 26% in the pre-treatment period to 9% with ivosidenib (108).
Recently, breakthrough results were achieved in this setting due to the use of another anti-IDH drug, vorasidenib. As a result, the phase III trial INDIGO may definitely change the therapeutic approach of low-grade astrocytomas. In this trial, patients with residual or recurrent grade 2 IDH-mutant glioma who had undergone no previous treatment other than surgery were randomly assigned patients to receive either an anti-IDH small molecule, oral vorasidenib or placebo. PFS was significantly improved in the vorasidenib group (mPFS 27.7 vs. 11.1 months) with a reduction of the risk of progression of 59% nearly. The time to the next intervention was significantly improved in the vorasidenib group (HR, 0.26; 95% CI: 0.15–0.43; P<0.001). AEs of grade 3 or higher occurred in 22.8% of the patients who received vorasidenib and in 13.5% of those who received placebo (109).
Hopefully, IDH-inhibitors will soon take part in clinical practice in neuro-oncology to completely change and improve life-expectancy of LGGs patients.
Discussion and future perspectives
As above mentioned, the biological importance of IDH alterations in cancer development is independent by the cell or cancer type and relies upon the overproduction of D-2HG and hypermethylation and blocks the normal differentiation patterns. In spite of these commonalities, the IDH-mutation activity differs in cancer subtypes on its role in metabolism and response to therapy, suggesting histologic-dependent, organ-dependent and even differentiation state-dependent phenotypes (110).
The knowledge of the biological effects of IDH mutation helps to find the correct setting amenable to reach the maximum efficacy after its inhibition.
First of all, the loss of wild-type or mIDH alleles leading to reduced D-2HG production during glioma progression, coupled with a greater efficacy to mIDH inhibitors in patients with non-enhancing gliomas, suggesting that targeting gliomas in their lower grade state, when most dependent on IDH mutations, must be considered for maximum therapeutic efficacy. As in the other oncological therapeutic strategies, the best first, and at an early stage (108,111).
Moreover, it is well known that immune response is downregulated in all IDH mutant cancer subtypes (112). Thus, IDH inhibitors could convert these immunologically cold tumors into immunologically hot tumors, improving the benefit of this therapeutic approach.
Lastly, we know that IDH mutations occur in the early stages of tumorigenesis, but several ongoing trials are enrolling patients with advanced-stage disease. Given that the dependence on IDH mutant metabolites varies between cancers and with tumour evolution, the timing of IDH-targeted therapy and the inclusion of additional agents are imperative to increase the outcomes of patients with IDH-mutated cancers. Unfortunately, the molecular search of this alteration is not feasible in all oncological centres. Thus, in addition to the rarity of IDH mutation, the lack of its standardised analysis may provoke an underestimation of its incidence.
Conclusions
In conclusion, great advances have been achieved in understanding the biology of IDH mutations in a variety of cancers. On the contrary, the pathogenic roles of this molecular alteration are not well understood. Certainly, these hotspot mutations remain a promising therapeutic target; hopefully, will dramatically change the therapeutic approach in some “orphan” tumors.
Acknowledgments
Funding: None.
Footnote
Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://cco.amegroups.com/article/view/10.21037/cco-24-17/rc
Peer Review File: Available at https://cco.amegroups.com/article/view/10.21037/cco-24-17/prf
Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://cco.amegroups.com/article/view/10.21037/cco-24-17/coif). R.A. serves as an unpaid editorial board member of Chinese Clinical Oncology from August 2022 to July 2024. The other authors have no conflicts of interest to declare.
Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.
Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.
References
- Borger DR, Tanabe KK, Fan KC, et al. Frequent mutation of isocitrate dehydrogenase (IDH)1 and IDH2 in cholangiocarcinoma identified through broad-based tumor genotyping. Oncologist 2012;17:72-9. [Crossref] [PubMed]
- Zhang Y, Wei H, Tang K, et al. Mutation analysis of isocitrate dehydrogenase in acute lymphoblastic leukemia. Genet Test Mol Biomarkers 2012;16:991-5. [Crossref] [PubMed]
- Mardis ER, Ding L, Dooling DJ, et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med 2009;361:1058-66. [Crossref] [PubMed]
- Parsons DW, Jones S, Zhang X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science 2008;321:1807-12. [Crossref] [PubMed]
- Paschka P, Schlenk RF, Gaidzik VI, et al. IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication. J Clin Oncol 2010;28:3636-43. [Crossref] [PubMed]
- Ward PS, Patel J, Wise DR, et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 2010;17:225-34. [Crossref] [PubMed]
- Yan H, Parsons DW, Jin G, et al. IDH1 and IDH2 mutations in gliomas. N Engl J Med 2009;360:765-73. [Crossref] [PubMed]
- Yang H, Ye D, Guan KL, et al. IDH1 and IDH2 mutations in tumorigenesis: mechanistic insights and clinical perspectives. Clin Cancer Res 2012;18:5562-71. [Crossref] [PubMed]
- Cairns G, Owen T, Kluzek S, et al. Therapeutic interventions in children and adolescents with patellar tendon related pain: a systematic review. BMJ Open Sport Exerc Med 2018;4:e000383. [Crossref] [PubMed]
- Chiang J, Harreld JH, Tinkle CL, et al. A single-center study of the clinicopathologic correlates of gliomas with a MYB or MYBL1 alteration. Acta Neuropathol 2019;138:1091-2. [Crossref] [PubMed]
- Gaal J, Burnichon N, Korpershoek E, et al. Isocitrate dehydrogenase mutations are rare in pheochromocytomas and paragangliomas. J Clin Endocrinol Metab 2010;95:1274-8. [Crossref] [PubMed]
- Kang MR, Kim MS, Oh JE, et al. Mutational analysis of IDH1 codon 132 in glioblastomas and other common cancers. Int J Cancer 2009;125:353-5. [Crossref] [PubMed]
- Lopez GY, Reitman ZJ, Solomon D, et al. IDH1(R132) mutation identified in one human melanoma metastasis, but not correlated with metastases to the brain. Biochem Biophys Res Commun 2010;398:585-7. [Crossref] [PubMed]
- Lang F, Jha A, Meuter L, et al. Identification of Isocitrate Dehydrogenase 2 (IDH2) Mutation in Carotid Body Paraganglioma. Front Endocrinol (Lausanne) 2021;12:731096. [Crossref] [PubMed]
- Venneker S, Bovée JVMG. IDH Mutations in Chondrosarcoma: Case Closed or Not? Cancers (Basel) 2023;15:3603. [Crossref] [PubMed]
- Solomou G, Finch A, Asghar A, et al. Mutant IDH in Gliomas: Role in Cancer and Treatment Options. Cancers (Basel) 2023;15:2883. [Crossref] [PubMed]
- Zhang Y, Liu Y, Lang F, et al. IDH mutation and cancer stem cell. Essays Biochem 2022;66:413-22. [Crossref] [PubMed]
- Internò V, Triggiano G, De Santis P, et al. Molecular Aberrations Stratify Grade 2 Astrocytomas Into Several Rare Entities: Prognostic and Therapeutic Implications. Front Oncol 2022;12:866623. [Crossref] [PubMed]
- Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol 2016;131:803-20. [Crossref] [PubMed]
- Radaelli E, Ceruti R, Patton V, et al. Immunohistopathological and neuroimaging characterization of murine orthotopic xenograft models of glioblastoma multiforme recapitulating the most salient features of human disease. Histol Histopathol 2009;24:879-91. [PubMed]
- Zeng W, Tang Z, Li Y, et al. Patient-derived xenografts of different grade gliomas retain the heterogeneous histological and genetic features of human gliomas. Cancer Cell Int 2020;20:1. [Crossref] [PubMed]
- Verheul C, Ntafoulis I, Kers TV, et al. Generation, characterization, and drug sensitivities of 12 patient-derived IDH1-mutant glioma cell cultures. Neurooncol Adv 2021;3:vdab103. [Crossref] [PubMed]
- Lee S, Urman A, Desai P. Emerging drug profile: Krebs cycle and cancer: IDH mutations and therapeutic implications. Leuk Lymphoma 2019;60:2635-45. [Crossref] [PubMed]
- Vienot A, Neuzillet C. Cholangiocarcinoma: the quest for a second-line systemic treatment. Transl Cancer Res 2019;8:S275-88. [Crossref] [PubMed]
- Bleeker FE, Atai NA, Lamba S, et al. The prognostic IDH1(R132) mutation is associated with reduced NADP+-dependent IDH activity in glioblastoma. Acta Neuropathol 2010;119:487-94. [Crossref] [PubMed]
- Dang L, White DW, Gross S, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 2009;462:739-44. [Crossref] [PubMed]
- Xu X, Zhao J, Xu Z, et al. Structures of human cytosolic NADP-dependent isocitrate dehydrogenase reveal a novel self-regulatory mechanism of activity. J Biol Chem 2004;279:33946-57. [Crossref] [PubMed]
- Adeva J. Current development and future perspective of IDH1 inhibitors in cholangiocarcinoma. Liver Cancer International 2022;3:17-31. [Crossref]
- Montalban-Bravo G, DiNardo CD. The role of IDH mutations in acute myeloid leukemia. Future Oncol 2018;14:979-93. [Crossref] [PubMed]
- Losman JA, Kaelin WG Jr. What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer. Genes Dev 2013;27:836-52. [Crossref] [PubMed]
- Xu W, Yang H, Liu Y, et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell 2011;19:17-30. [Crossref] [PubMed]
- Chowdhury R, Yeoh KK, Tian YM, et al. The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep 2011;12:463-9. [Crossref] [PubMed]
- Losman JA, Koivunen P, Kaelin WG Jr. 2-Oxoglutarate-dependent dioxygenases in cancer. Nat Rev Cancer 2020;20:710-26. [Crossref] [PubMed]
- Turcan S, Rohle D, Goenka A, et al. IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature 2012;483:479-83. [Crossref] [PubMed]
- Lu C, Ward PS, Kapoor GS, et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature 2012;483:474-8. [Crossref] [PubMed]
- Turcan S, Makarov V, Taranda J, et al. Mutant-IDH1-dependent chromatin state reprogramming, reversibility, and persistence. Nat Genet 2018;50:62-72. [Crossref] [PubMed]
- Sasaki M, Knobbe CB, Munger JC, et al. IDH1(R132H) mutation increases murine haematopoietic progenitors and alters epigenetics. Nature 2012;488:656-9. [Crossref] [PubMed]
- Tahiliani M, Koh KP, Shen Y, et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 2009;324:930-5. [Crossref] [PubMed]
- Figueroa ME, Abdel-Wahab O, Lu C, et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell 2010;18:553-67. [Crossref] [PubMed]
- Medeiros BC, Fathi AT, DiNardo CD, et al. Isocitrate dehydrogenase mutations in myeloid malignancies. Leukemia 2017;31:272-81. [Crossref] [PubMed]
- Waitkus MS, Diplas BH, Yan H. Isocitrate dehydrogenase mutations in gliomas. Neuro Oncol 2016;18:16-26. [Crossref] [PubMed]
- DiNardo CD, Propert KJ, Loren AW, et al. Serum 2-hydroxyglutarate levels predict isocitrate dehydrogenase mutations and clinical outcome in acute myeloid leukemia. Blood 2013;121:4917-24. [Crossref] [PubMed]
- Jalbert LE, Elkhaled A, Phillips JJ, et al. Metabolic Profiling of IDH Mutation and Malignant Progression in Infiltrating Glioma. Sci Rep 2017;7:44792. [Crossref] [PubMed]
- Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood 2017;130:722-31. [Crossref] [PubMed]
- Andronesi OC, Rapalino O, Gerstner E, et al. Detection of oncogenic IDH1 mutations using magnetic resonance spectroscopy of 2-hydroxyglutarate. J Clin Invest 2013;123:3659-63. [Crossref] [PubMed]
- Capper D, Weissert S, Balss J, et al. Characterization of R132H mutation-specific IDH1 antibody binding in brain tumors. Brain Pathol 2010;20:245-54. [Crossref] [PubMed]
- Boisselier B, Gállego Pérez-Larraya J, Rossetto M, et al. Detection of IDH1 mutation in the plasma of patients with glioma. Neurology 2012;79:1693-8. [Crossref] [PubMed]
- Mondesir J, Willekens C, Touat M, et al. IDH1 and IDH2 mutations as novel therapeutic targets: current perspectives. J Blood Med 2016;7:171-80. [Crossref] [PubMed]
- Duncan CG, Barwick BG, Jin G, et al. A heterozygous IDH1R132H/WT mutation induces genome-wide alterations in DNA methylation. Genome Res 2012;22:2339-55. [Crossref] [PubMed]
- Kernytsky A, Wang F, Hansen E, et al. IDH2 mutation-induced histone and DNA hypermethylation is progressively reversed by small-molecule inhibition. Blood 2015;125:296-303. [Crossref] [PubMed]
- Noushmehr H, Weisenberger DJ, Diefes K, et al. Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 2010;17:510-22. [Crossref] [PubMed]
- Saha SK, Parachoniak CA, Ghanta KS, et al. Mutant IDH inhibits HNF-4α to block hepatocyte differentiation and promote biliary cancer. Nature 2014;513:110-4. [Crossref] [PubMed]
- Wang F, Travins J, DeLaBarre B, et al. Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science 2013;340:622-6. [Crossref] [PubMed]
- Carbonneau M, M, Gagné L, Lalonde ME, et al. The oncometabolite 2-hydroxyglutarate activates the mTOR signalling pathway. Nat Commun 2016;7:12700. [Crossref] [PubMed]
- Karpel-Massler G, Ishida CT, Bianchetti E, et al. Induction of synthetic lethality in IDH1-mutated gliomas through inhibition of Bcl-xL. Nat Commun 2017;8:1067. [Crossref] [PubMed]
- Koivunen P, Lee S, Duncan CG, et al. Transformation by the (R)-enantiomer of 2-hydroxyglutarate linked to EGLN activation. Nature 2012;483:484-8. [Crossref] [PubMed]
- Losman JA, Looper RE, Koivunen P, et al. (R)-2-hydroxyglutarate is sufficient to promote leukemogenesis and its effects are reversible. Science 2013;339:1621-5. [Crossref] [PubMed]
- Sasaki M, Knobbe CB, Itsumi M, et al. D-2-hydroxyglutarate produced by mutant IDH1 perturbs collagen maturation and basement membrane function. Genes Dev 2012;26:2038-49. [Crossref] [PubMed]
- Bardella C, Al-Dalahmah O, Krell D, et al. Expression of Idh1(R132H) in the Murine Subventricular Zone Stem Cell Niche Recapitulates Features of Early Gliomagenesis. Cancer Cell 2016;30:578-94. [Crossref] [PubMed]
- Pirozzi CJ, Carpenter AB, Waitkus MS, et al. Mutant IDH1 Disrupts the Mouse Subventricular Zone and Alters Brain Tumor Progression. Mol Cancer Res 2017;15:507-20. [Crossref] [PubMed]
- Waitkus MS, Diplas BH, Yan H. Biological Role and Therapeutic Potential of IDH Mutations in Cancer. Cancer Cell 2018;34:186-95. [Crossref] [PubMed]
- Molenaar RJ, Botman D, Smits MA, et al. Radioprotection of IDH1-Mutated Cancer Cells by the IDH1-Mutant Inhibitor AGI-5198. Cancer Res 2015;75:4790-802. [Crossref] [PubMed]
- Popovici-Muller J, Lemieux RM, Artin E, et al. Discovery of AG-120 (Ivosidenib): A First-in-Class Mutant IDH1 Inhibitor for the Treatment of IDH1 Mutant Cancers. ACS Med Chem Lett 2018;9:300-5. [Crossref] [PubMed]
- Rohle D, Popovici-Muller J, Palaskas N, et al. An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science 2013;340:626-30. [Crossref] [PubMed]
- Yen K, Travins J, Wang F, et al. AG-221, a First-in-Class Therapy Targeting Acute Myeloid Leukemia Harboring Oncogenic IDH2 Mutations. Cancer Discov 2017;7:478-93. [Crossref] [PubMed]
- Pusch S, Krausert S, Fischer V, et al. Pan-mutant IDH1 inhibitor BAY 1436032 for effective treatment of IDH1 mutant astrocytoma in vivo. Acta Neuropathol 2017;133:629-44. [Crossref] [PubMed]
- DiNardo CD, Schimmer AD, Yee KWL, et al. A Phase I study of IDH305 in patients with advanced malignancies including relapsed/refractory AML and MDS that harbor IDH1R132 Mutations. Blood 2016;128:1073. [Crossref]
- Machida Y, Matsumoto N, Isono T, et al. Phonon hydrodynamics and ultrahigh-room-temperature thermal conductivity in thin graphite. Science 2020;367:309-12. [Crossref] [PubMed]
- Caravella JA, Lin J, Diebold RB, et al. Structure-Based Design and Identification of FT-2102 (Olutasidenib), a Potent Mutant-Selective IDH1 Inhibitor. J Med Chem 2020;63:1612-23. [Crossref] [PubMed]
- Tey A, Schwarer J, Raffa R, et al. High risk of infection in 'real-world' patients receiving ibrutinib, idelalisib or venetoclax for mature B-cell leukaemia/lymphoma. Eur J Haematol 2023;110:540-7. [Crossref] [PubMed]
- Amatangelo MD, Quek L, Shih A, et al. Enasidenib induces acute myeloid leukemia cell differentiation to promote clinical response. Blood 2017;130:732-41. [Crossref] [PubMed]
- Intlekofer AM, Shih AH, Wang B, et al. Acquired resistance to IDH inhibition through trans or cis dimer-interface mutations. Nature 2018;559:125-9. [Crossref] [PubMed]
- Zhuang X, Pei HZ, Li T, et al. The Molecular Mechanisms of Resistance to IDH Inhibitors in Acute Myeloid Leukemia. Front Oncol 2022;12:931462. [Crossref] [PubMed]
- Choe S, Wang H, DiNardo CD, et al. Molecular mechanisms mediating relapse following ivosidenib monotherapy in IDH1-mutant relapsed or refractory AML. Blood Adv 2020;4:1894-905. [Crossref] [PubMed]
- Jung YS, Park JI. Wnt signaling in cancer: therapeutic targeting of Wnt signaling beyond β-catenin and the destruction complex. Exp Mol Med 2020;52:183-91. [Crossref] [PubMed]
- Kaufman JL, Gasparetto C, Schjesvold FH, et al. Targeting BCL-2 with venetoclax and dexamethasone in patients with relapsed/refractory t(11;14) multiple myeloma. Am J Hematol 2021;96:418-27. [Crossref] [PubMed]
- Harding JJ, Lowery MA, Shih AH, et al. Isoform Switching as a Mechanism of Acquired Resistance to Mutant Isocitrate Dehydrogenase Inhibition. Cancer Discov 2018;8:1540-7. [Crossref] [PubMed]
- Chan SM, Thomas D, Corces-Zimmerman MR, et al. Isocitrate dehydrogenase 1 and 2 mutations induce BCL-2 dependence in acute myeloid leukemia. Nat Med 2015;21:178-84. [Crossref] [PubMed]
- Lachowiez CA, Borthakur G, Loghavi S, et al. Phase Ib/II study of the IDH1-mutant inhibitor ivosidenib with the BCL2 inhibitor venetoclax +/- azacitidine in IDH1-mutated hematologic malignancies. J Clin Oncol 2020;38:7500. [Crossref]
- Gil-Sierra MD, Briceño-Casado MP, Sierra-Sanchez JF. Ivosidenib and Azacitidine in IDH1-Mutated AML. N Engl J Med 2022;386:2535-6. [Crossref] [PubMed]
- Razumilava N, Gores GJ. Liver transplantation for intrahepatic cholangiocarcinoma--Authors' reply. Lancet 2014;384:1182-3. [Crossref] [PubMed]
- Banales JM, Marin JJG, Lamarca A, et al. Cholangiocarcinoma 2020: the next horizon in mechanisms and management. Nat Rev Gastroenterol Hepatol 2020;17:557-88. [Crossref] [PubMed]
- Xiang X, Liu Z, Zhang C, et al. IDH Mutation Subgroup Status Associates with Intratumor Heterogeneity and the Tumor Microenvironment in Intrahepatic Cholangiocarcinoma. Adv Sci (Weinh) 2021;8:e2101230. [Crossref] [PubMed]
- Zhang W, Zhou H, Wang Y, et al. Systemic treatment of advanced or recurrent biliary tract cancer. Biosci Trends 2020;14:328-41. [Crossref] [PubMed]
- Grassian AR, Pagliarini R, Chiang DY. Mutations of isocitrate dehydrogenase 1 and 2 in intrahepatic cholangiocarcinoma. Curr Opin Gastroenterol 2014;30:295-302. [Crossref] [PubMed]
- Boscoe AN, Rolland C, Kelley RK. Frequency and prognostic significance of isocitrate dehydrogenase 1 mutations in cholangiocarcinoma: a systematic literature review. J Gastrointest Oncol 2019;10:751-65. [Crossref] [PubMed]
- Pirozzi CJ, Yan H. The implications of IDH mutations for cancer development and therapy. Nat Rev Clin Oncol 2021;18:645-61. [Crossref] [PubMed]
- Ding N, Che L, Li XL, et al. Oncogenic potential of IDH1R132C mutant in cholangiocarcinoma development in mice. World J Gastroenterol 2016;22:2071-80. [Crossref] [PubMed]
- Wu MJ, Shi L, Dubrot J, et al. Mutant IDH Inhibits IFNγ-TET2 Signaling to Promote Immunoevasion and Tumor Maintenance in Cholangiocarcinoma. Cancer Discov 2022;12:812-35. [Crossref] [PubMed]
- Rizzo A, Ricci AD, Bonucci C, et al. Experimental HER2- targeted therapies for biliary tract cancer. Expert Opin Investig Drugs 2021;30:389-99. [Crossref] [PubMed]
- Ludwig H, Delforge M, Facon T, et al. Prevention and management of adverse events of Novel agents in multiple myeloma: A consensus of the european myeloma network. Leukemia 2017; Epub ahead of print. [Crossref] [PubMed]
- Bose P, Vachhani P, Cortes JE. Treatment of Relapsed/Refractory Acute Myeloid Leukemia. Curr Treat Options Oncol 2017;18:17. [Crossref] [PubMed]
- Miller JJ, Shih HA, Andronesi OC, et al. Isocitrate dehydrogenase-mutant glioma: Evolving clinical and therapeutic implications. Cancer 2017;123:4535-46. [Crossref] [PubMed]
- Jusakul A, Cutcutache I, Yong CH, et al. Whole-Genome and Epigenomic Landscapes of Etiologically Distinct Subtypes of Cholangiocarcinoma. Cancer Discov 2017;7:1116-35. [Crossref] [PubMed]
- Wang ML, Rule S, Martin P, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med 2013;369:507-16. [Crossref] [PubMed]
- Lowery MA, Burris HA 3rd, Janku F, et al. Safety and activity of ivosidenib in patients with IDH1-mutant advanced cholangiocarcinoma: a phase 1 study. Lancet Gastroenterol Hepatol 2019;4:711-20. [Crossref] [PubMed]
- Abou-Alfa GK, Macarulla T, Javle MM, et al. Ivosidenib in IDH1-mutant, chemotherapy-refractory cholangiocarcinoma (ClarIDHy): a multicentre, randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol 2020;21:796-807. Erratum in: Lancet Oncol 2020;21:e462; Lancet Oncol 2024;25:e61. [Crossref] [PubMed]
- Saha SK, Gordan JD, Kleinstiver BP, et al. Isocitrate Dehydrogenase Mutations Confer Dasatinib Hypersensitivity and SRC Dependence in Intrahepatic Cholangiocarcinoma. Cancer Discov 2016;6:727-39. [Crossref] [PubMed]
- Sharma N, Sisodia S, Jain A, et al. Evaluation of the Efficacy of Recent Caries Removal Techniques: An In Vitro Study. Cureus 2023;15:e34432. [Crossref] [PubMed]
- Zhu AX, Macarulla T, Javle MM, et al. Final Overall Survival Efficacy Results of Ivosidenib for Patients With Advanced Cholangiocarcinoma With IDH1 Mutation: The Phase 3 Randomized Clinical ClarIDHy Trial. JAMA Oncol 2021;7:1669-77. [Crossref] [PubMed]
- Rizzo A, Ricci AD, Brandi G. IDH inhibitors in advanced cholangiocarcinoma: Another arrow in the quiver? Cancer Treat Res Commun 2021;27:100356. [Crossref] [PubMed]
- Cancer Genome Atlas Research Network. Comprehensive, Integrative Genomic Analysis of Diffuse Lower-Grade Gliomas. N Engl J Med 2015;372:2481-98. [Crossref] [PubMed]
- Verhaak RG, Hoadley KA, Purdom E, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010;17:98-110. [Crossref] [PubMed]
- Ceccarelli M, Barthel FP, Malta TM, et al. Molecular Profiling Reveals Biologically Discrete Subsets and Pathways of Progression in Diffuse Glioma. Cell 2016;164:550-63. [Crossref] [PubMed]
- Grabowski MM, Sankey EW, Ryan KJ, et al. Immune suppression in gliomas. J Neurooncol 2021;151:3-12. [Crossref] [PubMed]
- Kohanbash G, Carrera DA, Shrivastav S, et al. Isocitrate dehydrogenase mutations suppress STAT1 and CD8+ T cell accumulation in gliomas. J Clin Invest 2017;127:1425-37. [Crossref] [PubMed]
- Amankulor NM, Kim Y, Arora S, et al. Mutant IDH1 regulates the tumor-associated immune system in gliomas. Genes Dev 2017;31:774-86. [Crossref] [PubMed]
- Arvanitis CD, Ferraro GB, Jain RK. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat Rev Cancer 2020;20:26-41. [Crossref] [PubMed]
- Mellinghoff IK, Ellingson BM, Touat M, et al. Ivosidenib in Isocitrate Dehydrogenase 1-Mutated Advanced Glioma. J Clin Oncol 2020;38:3398-406. [Crossref] [PubMed]
- Mellinghoff IK, van den Bent MJ, Blumenthal DT, et al. Vorasidenib in IDH1- or IDH2-Mutant Low-Grade Glioma. N Engl J Med 2023;389:589-601. [Crossref] [PubMed]
- Unruh D, Zewde M, Buss A, et al. Methylation and transcription patterns are distinct in IDH mutant gliomas compared to other IDH mutant cancers. Sci Rep 2019;9:8946. [Crossref] [PubMed]
- Mazor T, Chesnelong C, Pankov A, et al. Clonal expansion and epigenetic reprogramming following deletion or amplification of mutant IDH1. Proc Natl Acad Sci U S A 2017;114:10743-8. [Crossref] [PubMed]