Next Article in Journal
Therapeutic Effects of Curcumin Derivatives against Obesity and Associated Metabolic Complications: A Review of In Vitro and In Vivo Studies
Next Article in Special Issue
ULK2 Is a Key Pro-Autophagy Protein That Contributes to the High Chemoresistance and Disease Relapse in FLT3-Mutated Acute Myeloid Leukemia
Previous Article in Journal
Genomic Factors and Therapeutic Approaches in HIV-Associated Neurocognitive Disorders: A Comprehensive Review
Previous Article in Special Issue
DNA Repair Pathway in Ovarian Cancer Patients Treated with HIPEC
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Toward a Unifying Hypothesis for Redesigned Lipid Catabolism as a Clinical Target in Advanced, Treatment-Resistant Carcinomas

by
Paul M. Bingham
* and
Zuzana Zachar
Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(18), 14365; https://doi.org/10.3390/ijms241814365
Submission received: 31 July 2023 / Revised: 15 September 2023 / Accepted: 18 September 2023 / Published: 21 September 2023

Abstract

:
We review extensive progress from the cancer metabolism community in understanding the specific properties of lipid metabolism as it is redesigned in advanced carcinomas. This redesigned lipid metabolism allows affected carcinomas to make enhanced catabolic use of lipids in ways that are regulated by oxygen availability and is implicated as a primary source of resistance to diverse treatment approaches. This oxygen control permits lipid catabolism to be an effective energy/reducing potential source under the relatively hypoxic conditions of the carcinoma microenvironment and to do so without intolerable redox side effects. The resulting robust access to energy and reduced potential apparently allow carcinoma cells to better survive and recover from therapeutic trauma. We surveyed the essential features of this advanced carcinoma-specific lipid catabolism in the context of treatment resistance and explored a provisional unifying hypothesis. This hypothesis is robustly supported by substantial preclinical and clinical evidence. This approach identifies plausible routes to the clinical targeting of many or most sources of carcinoma treatment resistance, including the application of existing FDA-approved agents.

1. Introduction

Successful clinical targeting of carcinoma metabolism requires that we develop a relatively complete, coherent, predictive theory of how normal cell and carcinoma cell metabolism differ. Our goal here is narrow: to review the current status of an important element of this effort. We will endeavor to contextualize one specific area of progress: changes in the properties and regulation of lipid catabolism that are strongly associated with and apparently cause treatment resistance in advanced carcinomas.
We organize this discussion around an emerging provisional hypothesis describing the core features of this process. Sarcomas and liquid tumors can show some of the lipid metabolic features we focus on here, as we will briefly mention below. However, the story we review is largely based on the much more extensive body of data from carcinomas; we will largely focus throughout on this tumor class.
It is useful to begin with the longstanding insight that carcinomas are a particular class of deranged tissue. Specifically, carcinomas pervasively resemble healing wounds that never resolve to complete the normal healing process [1,2,3,4,5,6,7,8]. These insights strongly imply that features of carcinoma metabolism will reflect the mobilization and strategic modification of evolved mechanisms originally designed to support wound healing, as follows:
First is the reregulation of catabolism to deal with highly fluctuating and often inadequate access to nutrients and oxygen. Among these challenges is supporting catabolism sufficient for survival and ultimate growth without the generation of toxic levels of reactive oxygen species (ROS) [8,9,10,11].
The second is the reregulation of cell death decision-making. Cells in normal healing wounds need to make an adaptive choice between programmed cell death (commonly colloquially referred to as the “scab” response in wound healing literature) or survival, with retention of the capacity for growth and/or differentiation (the “scar” response). We expect the genetic and epigenetic evolution of carcinomas to mobilize (and refine) catabolic processes in support of growth but also to avoid the cell death responses that normal healing wound cells would undergo. Effective therapeutic assault must be sufficient to overcome this cell death resistance.
The key implications of this broader picture are as follows: Lipids are an especially important nutrient for stressed carcinoma cells and normal healing wound epithelial cells. This unique value of lipids results from their exceptional density and economy of storage of energy/reducing potential, and the crucial role of lipids as structural molecules in membranes and, occasionally, as signals [12,13,14]. The downside of lipids in the carcinoma/wound environment is that their exploitation for energy is very strongly dependent on oxygen, again, a resource commonly severely limited and variable in this context.
Thus, we expect the evolved wound healing machinery to be constructed to allow individual epithelial cells to control flux through lipid metabolism based on oxygen availability, both to support survival/growth behavior when oxygen is adequate and to trigger cell death when oxygen levels are too low to match lipid availability. In turn, as noted, we expect this machinery to be coopted and reprogrammed in the evolution of advanced malignancy, including attenuation of the cell death response.
A large body of work on lipid metabolism in advanced, treatment-resistant carcinomas fits well with this picture (reviewed below). Moreover, because the founding machinery is relatively exotic (wound healing), its modified version, driving broad treatment resistance in advanced carcinomas, is expected to have features that can be targeted in patients without unacceptable side effects.
The following section is a high-altitude overview of the provisional hypothesis we will explore.

2. Context of “Lipid Metabolism Resistance System” (LMRS) Hypothesis

Over the last decade, it has become increasingly clear that changes in lipid metabolism are commonly associated with advanced, treatment-resistant carcinomas [15,16,17,18,19,20,21,22,23,24,25,26,27,28]. There is extensive evidence for ongoing redesign of lipid metabolism in tumor cells as they progress to more advanced, treatment-resistant carcinoma [29,30,31,32,33,34,35,36]. Moreover, hypoxia, commonly experienced by carcinoma cells in vivo and representing a negative clinical prognostic, upregulates fatty acid uptake and storage [37,38,39]. Hypoxia interacts with ROS production in various ways beyond those we will discuss in detail below. These interactions include those mediated by known regulators, such as HIF1 and others [40,41,42,43,44,45].
Finally, clonal selection processes operate on carcinoma stem cells in tumors [46]. These processes generate cell subpopulations wherein lipid storage/catabolism upregulation and ensuing treatment resistance become characteristic of the advanced carcinomas they found.
While other features of carcinoma metabolism might also be of therapeutic interest [47,48,49,50], our primary focus here is the putatively universal core features of lipid catabolism specifically involved in producing treatment-resistant carcinomas (Figure 1).
We will refer to this provisional picture as the “lipid metabolism resistance system” or LMRS (Figure 1). One of the central assumptions of this hypothesis is that elevated lipid catabolism provides increased access to energy and reduces potential. These increases, in turn, are proposed to allow carcinoma cells to better survive and recover from diverse therapeutic assaults, thereby rendering them broadly treatment-resistant.
The precise biochemical or cellular role(s) of LMRS metabolism in supporting carcinoma cell survival, growth, and treatment resistance remains to be clearly characterized. At present, experimental data are sufficient to support this causal relationship but inadequate to yield a fully detailed mechanistic picture of the basis of this effect.
We review in detail below the steps in this specialized, resistance-associated catabolism of fatty acids in carcinoma cells. This redesign for elevated use and dependence upon lipid catabolism begins with generating the lipid substrates supporting this process. This frequently includes the elevated capacity of carcinoma cells to synthesize fatty acids de novo [16,51,52,53]. Also common is the enhanced carcinoma cell ability to actively import fatty acids and their multimeric composites from their surroundings [26,54,55,56,57,58]. Finally, carcinoma cells interact with and reciprocally manipulate surrounding stromal cells to procure elevated access to the lipids these support cells can manufacture and export [32,57,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76].
In spite of their importance, these diverse processes entail sufficient redundancy that attempting to clinically target individual components is likely to be ineffective and highly susceptible to evolved resistance. Thus, we will not emphasize elevated carcinoma cell lipid access further here. Rather, we will focus on the catabolic processing of fatty acids once they arrive in advanced, treatment-resistant carcinoma cells.

3. Detailed Description of the Steps in the LMRS Hypothesis

The LMRS process includes three major steps (Figure 1). First is peroxisomal fatty acid beta-oxidation, initiated by acyl-coenzyme A oxidases (Acox). Second, is desaturation, resulting in a monounsaturated fatty acid (MUFA) derived from saturated fatty acid precursors and commonly catalyzed by stearoyl-CoA desaturase (SCD1). This step may precede or follow Acox oxidation. Finally, the products of these first two steps are transferred to the mitochondrion for complete oxidation to CO2 and H2O.
It is crucial to recognize that all these steps are oxygen-dependent in ways that allow catabolic flux of fatty acids to be controlled on the basis of real-time oxygen availability.
We review each of these steps and its relevance to carcinoma treatment resistance in detail as below.

3.1. Mitochondrial Oxygen-Dependent Metabolism of Preprocessed Fatty Acids

The LMRS hypothesis is more easily grasped by beginning at the terminal mitochondrial steps. As synopsized above, the presumptive role of the preceding, extra-mitochondrial steps in fatty acid catabolism is to control flux in response to limited and potentially fluctuating oxygen availability (in wound healing or carcinoma microenvironments). Both peroxisomal beta-oxidation and desaturase processing require molecular oxygen, rendering these steps “licensed” and rate-controlled by oxygen availability (Section 3.2 and Section 3.3).
To grasp our provisional interpretation of this process, it is essential to understand how the final mitochondrial steps of fatty acid catabolism are, likewise, managed in response to low, variable oxygen levels. Oxygen regulation of LMRS fatty acid catabolism is pervasive from the beginning to the end of the process.
When oxygen availability is limited, several mitochondrial processes generate elevated levels of reactive oxygen species, ultimately or directly in the form of hydrogen peroxide, H2O2, as discussed in more detail below [77]. We note that other ROS species are produced in some of these processes, especially superoxide; however, these other ROS species are generally either rapidly destroyed or converted to H2O2. Moreover, H2O2 appears to be the primary informational ROS molecule in the cases we discuss [77]. Thus, it is reasonable to focus on hydrogen peroxide.
On the one hand, the mitochondrial electron transport complexes (ETC) retain elevated steady-state levels of electrons when these cannot be off-loaded at the end step in the ETC system (complex IV; cytochrome c oxidase) to form water from oxygen. Instead, these excess electrons are donated directly by intermediate ETC steps to whatever oxygen remains, creating H2O2 [77]. On the other hand, matrix oxoacid dehydrogenase complexes, especially alpha-ketoglutarate dehydrogenase (KGDH), also donate reducing potential to residual oxygen (again, making H2O2) when their NADH product levels become elevated in the absence of the ability for NADH electrons to be efficiently passed off to a now-saturated ETC [78,79,80,81].
Mitochondrial metabolism is expected to have evolved to control this hypoxia-driven elevated ROS generation. Such control appears to be reflected in the adaptive redox regulation of several steps through H2O2-dependent modification. This redox blockade has the overall effect of suppressing further fatty acid flux in the face of oxygen deprivation-dependent clogging of mitochondrial metabolism (and, thus, increased H2O2 production).
On the one hand, elevated mitochondrial matrix H2O2 levels suppress further fatty acid import. This includes redox modification of the matrix face of the CAC transporter (mitochondrial carnitine acyl-carnitine carrier, Figure 1; SLC25A20), which blocks fatty acyl coenzyme A thioester import across the inner mitochondrial membrane [82].
On the other hand, several rate-limiting steps in the TCA cycle oxidation of fatty acid-derived acetate units are, likewise, highly sensitive to redox inhibition in the presence of elevated H2O2 levels [83]. These include aconitase [84] and KGDH [78,79,80,81,85] (Figure 1). Notably, H2O2 produced by KGDH constitutes a local negative feedback loop controlling this enzyme itself, in addition to contributing more broadly to the determination of regulatory levels of matrix redox signals [78,79]. These redox inhibition steps choke off further fatty acid-dependent electron generation when oxygen levels are insufficient.
As discussed in Castelli et al. [42,86,87] and Ying and Hu [88], mitochondrial fatty acid oxidation (FAO) can generate elevated levels of reactive oxygen species (ROS), while, reciprocally, driving antioxidant control of ROS. The relative rates of these two opposing processes are strongly influenced by oxygen availability, as described below.
There are various mitochondrial enzymatic systems for generating the antioxidant capacity needed to manage/eliminate metabolism-generated ROS. Among the most important of these is the transhydrogenase complex (NNT) [81,89,90,91,92]. The NNT complex spans the inner mitochondrial membrane and exploits energy stored in the Mitchell/Moyle proton gradient to drive the transfer of reducing potential from NADH to NADPH. Major mitochondrial antioxidant systems commonly require NADPH as a direct or ultimate source of reducing potential [77]. When oxygen levels are limited, one of the drivers of the NNT forward reaction, the proton gradient, will be attenuated as a result of the clogged electron transport through the inner membrane ETC hydrogen ion pumps. This effect, in turn, depresses levels of NADP+ reduction to NADPH and, thus, further enhances steady-state H2O2 levels (again, in turn, lowering catabolic flux through the TCA cycle as discussed above).
NNT control of mitochondrial matrix redox levels is especially important in LMRS catabolism. Specifically, NNT is potently inhibited by saturated fatty acid CoA thioesters, the primary matrix form of these fatty acids [93,94,95]. Thus, if levels of saturated fatty acids (especially palmitate and stearate) are elevated in the matrix as a result of desaturation failure in the absence of adequate cytosolic oxygen levels (Section 3.3), mitochondrial matrix H2O2 levels are expected to show an additional increase, suppressing further fatty acid import and catabolism (above).
In view of these roles in managing oxygen-limited mitochondrial metabolism, it is plausible that the NNT system will be reregulated in advanced carcinomas, as indicated below.
Illuminating is the recent work of Han et al., who showed that elevated NNT enzymatic activity enhanced gastric carcinoma mitochondrial metabolism and resistance to immunotherapy [96]. Moreover, an upstream trigger for the post-translational modification producing this elevated NNT activity is IL-1beta. This trigger, in turn, is sufficient to drive local inflammation and, ultimately, gastric carcinoma. Thus, it is reasonable to anticipate that this inflammation/carcinoma-dependent activation of NNT might be broadly general.
Further supporting this picture, genetic ablation of NNT interferes with malignancy in preclinical systems, including adrenal carcinoma [97], renal carcinoma [98], gastric carcinoma [99], and nonsmall cell lung carcinoma (NSCLC) [100].
Notably, KGDH and NNT appear to interact intimately (perhaps directly), as evidenced by their coparticipation in reductive mitochondrial citrate synthesis [101]. Moreover, there is circumstantial evidence that KGDH redox regulatory processes are modified in carcinoma cells [78].
Finally, NNT reregulation in carcinomas occasionally involves downregulation rather than the more common upregulation discussed above [102,103]. It remains to be seen what the implications of such minority observations are for the generality of the LMRS hypothesis in its simplest form.

3.2. Acox-Catalyzed, Oxygen-Dependent Peroxisomal Fatty Acid Beta-Oxidation

There is substantial evidence that peroxisomes interact with mitochondria, including through direct contact, and thereby contribute to the control of mitochondrial lipid metabolism [104,105,106,107,108]. Moreover, these peroxisomal/mitochondrial interactions are directly implicated in determining carcinoma lipid metabolism [109,110]. Especially notable here, elevated expression of mitochondrial/peroxisomal interaction mediators has negative clinical prognostic implications for both sarcomas and carcinomas [111].
Sarcomas yield a particularly useful insight. Elevation of one of these peroxisomal/mitochondrial interaction regulators (phosphatidylinositol-5-phosphate 4-kinase type 2 alpha; PI5P4Kalpha) is also correlated with advanced, aggressive status in sarcomas (op cit.). Moreover, PI5P4Kalpha knockout dramatically reduced KRAS/p53KO-dependent sarcoma formation in a mouse system and the growth of in vivo tumors from cell lines derived from these primary tumors (op cit.).
The beta-oxidation of fatty acids in peroxisomes is of special interest for clinical targeting. Specifically, peroxisomal beta-oxidation is very commonly essential for advanced, treatment-resistant carcinoma cells (below). In contrast, differentiated normal cells appear to rely largely or entirely on direct mitochondria-autonomous uptake systems to support fatty acid oxidation [112].
Unlike the analogous mitochondrial process, peroxisomal fatty acid beta-oxidation is initiated by an oxidase reaction that directly burns an oxygen molecule. This process transfers the abstracted electrons to oxygen with the production of hydrogen peroxide. This H2O2 is then rapidly destroyed by catalase in the peroxisomal matrix. This initial fatty acid reaction is catalyzed by the Acox oxidases. This process has the effect of licensing the ultimate transfer of shortened fatty acids and acetate units to the mitochondrion on the basis of the presence of adequate levels of molecular oxygen to carry out the oxidase reaction [109,113].
Several studies robustly support a central role for peroxisomal beta-oxidation of fatty acids in advanced, treatment-resistant carcinomas, as predicted by the LMRS hypothesis.
First, Cai et al. showed that peroxisomes were essential to liver carcinoma cell survival in vitro and xenograft growth in vivo [114]. Remarkably, the same gene knockout/knockdown (PEX2) that eliminates peroxisome function in these carcinoma cell studies nonetheless permits normal embryonic development. This observation directly supports the other evidence discussed herein that the pattern of peroxisomal metabolism seen in carcinomas is preferentially important to these cancers.
Second, a later step in the peroxisomal fatty acid beta-oxidation pathway generates reduced NADH. Unlike the FADH2 intermediate generated by the first Acox step, these electrons cannot be transferred to molecular oxygen for H2O2 production. Instead, NAD+ must be regenerated through mitochondrial shuttles, most importantly a lactate/pyruvate shuttle. This shuttle appears to depend on the monocarboxylate transporter isoform, MCT2. MCT2 is a high-affinity transporter of pyruvate and lactate across membrane bilayers. Unlike other members of this transporter family that are localized to the plasma membrane, MCT2 commonly localizes to the peroxisomal membrane (and probably also the mitochondrial membrane) [115]. Crucially, in this context, Valenca et al. have shown that MCT2 is upregulated in prostate carcinoma and that peroxisomal MCT2 localization is essential to prostate carcinoma cell growth [116,117].
Of particular relevance to the LMRS hypothesis, this MCT2 shuttle is limited by oxygen-dependent mitochondrial disposal of electrons from lactate conversion back to pyruvate [118]. Thus, this peroxisomal shuttle shares the feature of oxygen availability-licensing of lipid catabolism with the Acox oxidase and desaturase reactions (Section 3.3 below).
Third, BRAF mutant melanoma cells generate cell subclones, dubbed persisters, that are resistant to clinical drug targeting of this kinase. These persister cells are thought to be the major source of clinical drug resistance. Shen et al. showed that these drug-resistant cells are dependent on peroxisomal fatty acid oxidation [119]. Genetic knockdown or drug inhibition of the peroxisomal Acox1 oxidase eliminates these persister cells.
Fourth, the carnitine O-octanoyltransferase (CROT) and carnitine acetyltransferase (CRAT) transporters are responsible for moving shortened fatty acid and acetate units from peroxisomal fatty acid beta-oxidation to the mitochondrion for completion of the oxidation process. Lasheras-Otero et al. showed that these transporters are essential for the survival of detached melanoma cells in vitro (proxies for circulating carcinoma cells in vivo) and that suppression of CROT or CRAT substantially reduces melanoma metastasis in in vivo preclinical models [120]. Moreover, these effects of CROT/CRAT knockdown can be mimicked with a small-molecule Acox oxidase inhibitor (op cit.).
Fifth, our group characterized carcinoma resistance to the CPI-613 tumor-specific TCA cycle inhibitor [121]. We showed that in vitro resistance to this drug depended on elevated accumulation and catabolism of lipid stores. We were able to block this resistance with the small-molecule Acox inhibitor, thioridazine. Moreover, thioridazine was also potent in sensitizing in vivo xenografts of an otherwise CPI-613-resistant pancreatic carcinoma (PDAC) cell line (AsPC1; op cit.). In vitro studies indicated that CPI-613 resistance was dependent on peroxisomal beta-oxidation in every carcinoma cell type tested. Further, inhibitor studies implicate acute MET kinase signaling in maintaining this LMRS lipid-dependent peroxisomal rescue pathway. The FDA-approved MET inhibitor (crizotinib; CRZ) sensitized resistant AsPC1 PDAC xenografts analogously to thioridazine (op cit.).
A substantial body of evidence further implies an important role of peroxisomal fatty acid catabolism in carcinoma treatment resistance. In spite of the reduction in total amounts/numbers of peroxisomes in several carcinoma cases [122,123], there is also a common observation of an elevation in specific peroxisomal enzymatic activities in various cancers, including carcinomas.
Dahabieh et al. showed that the upregulation of peroxisomal enzymes supports lymphoma cell resistance to an HDAC inhibitor anticancer agent [124]. Further, Dahabieh et al. showed that reducing peroxisome levels through stimulation of plexophagy sensitized resistant lymphoma cells to this HDAC inhibitor [125]. Zheng et al. showed that Acox1 levels determined doxorubicin sensitivity in lymphoma cells [126].
Kim et al. found that strong Acox1 expression was a significant negative clinical prognostic for breast carcinoma [127]. Yu et al. showed that Acox activity supports prostate carcinoma cell proliferation, migration, and invasion in vitro [128]. Kuna et al. showed that peroxisomal beta-oxidation can be a robust source of fatty acid catabolism in an NSCLC carcinoma cell line [129].
Tamatani et al. [130] and Okamoto et al. [131] showed that Acox1 overexpression can exhibit oncogene-like activity, permitting in vivo xenograft carcinoma formation by otherwise nonmalignant cells. These striking results indicate that the elevation of LMRS-like lipid metabolism is sufficient to robustly reduce barriers to carcinoma formation.
Finally, genetic ablation of peroxisomes in Drosophila produces lethality at the pupal stage. Larval development happens normally, irrespective of peroxisome knockout. Pupal development involves massive histolysis of larval tissues, followed by extensive stem cell division. It is during these processes that the peroxisomal knockout produces lethality. Thus, pupal development likely makes use of wound-healing machinery, and this process is significantly and selectively compromised without peroxisomes [132].
These authors made a further striking observation. They showed that these consequences of peroxisome loss in Drosophila can be largely eliminated by providing a mixture of saturated and unsaturated medium-chain-length fatty acids in the diet. In context, these results indicate that the pupal developmental requirement for peroxisomes can be replaced by the products of peroxisomal beta-oxidation, the shortening of fatty acid chain length. Further consistent with this view, the loss of mammalian melanoma stem cell populations through peroxisomal attenuation can be rescued by comparable medium-chain-length fatty acid supplementation [120]. In aggregate, these data support the picture that beta-oxidation-dependent provision of shortened fatty acids is the peroxisomal process sustaining clinically relevant carcinoma cell behavior, consistent with the proposed details of the LMRS hypothesis (Figure 1).

3.3. Oxygen-Dependent Fatty Acid Desaturation by the SCD1 Enzyme Anchored on the Cytoplasmic Face of the Endoplamic Reticulum (ER)

Stearoyl-coenzyme A desaturase 1 (SCD1) is the most thoroughly characterized component of LMRS (Figure 1), providing the most robust and extensive opportunities to test this hypothesis.
Key evidence indicates that SCD1 participates in normal tissue wound healing, as the LMRS hypothesis predicts [133,134]. Likewise, the well-defined mouse adipocyte-supported hair follicle regeneration/wound healing system is dependent on SCD1 [135]. Also striking is the extent to which SCD1 appears to be actively involved in the control of metabolism in diverse normal cell settings [136].
SCD1 catalyzes the introduction of a double bond (desaturation) into saturated fatty acids from endogenous lipogenesis or exogenous sources. SCD1 substrates commonly include palmitate (C16:0) and stearate (C18:0), resulting in their conversion to palmitoleate (C16:1 n-7) and oleate (C18:1 n-8), respectively [137,138,139]. Among its diverse effects, this desaturation is expected to prevent saturated fatty acid inhibition of mitochondrial NNT and, thus, prevent ensuing inhibition of mitochondrial metabolism (Section 3.1).
The striking features of SCD1 function are two. First, as noted, this reaction makes direct use of oxygen and is inhibited at low oxygen levels [137,138,140]. Second, SCD1 expression is crucial to the function and survival of normal stem cells (above) and is commonly upregulated in carcinomas in general. This SCD1 upregulation is often especially extreme in carcinoma stem cells (CSCs), which are thought to be responsible for most treatment resistance. This work has been well discussed recently [141,142,143,144,145,146]. Our focus here will be on the issues most directly relevant to the provisional LMRS hypothesis.
Cases where SCD1 expression and/or upregulation are crucial in advanced malignancy, including in treatment resistance, are diverse. The following are illuminating examples: Scaglia and Igal [147] and Scaglia et al. [148] showed that SCD1 plays a crucial role in tumor cell proliferation and metabolism in transformed fibroblasts and several carcinoma cell models, including NSCLC. Xuan et al. showed that SCD1 and a related oxidase are essential to malignancy and platinum drug resistance in an ovarian carcinoma model [149]. Huang et al. found that SCD1 upregulation is associated with late-stage lung carcinoma, representing a poor clinical prognostic indicator [150]. Pisanu et al. showed that SCD1 expression is necessary for cisplatin resistance in lung carcinoma stem cells and that elevated SCD1 expression is a negative prognostic in lung carcinoma patients [151]. Pisanu et al. showed that the evolved resistance of melanomas to BRAF inhibitors depends on SCD1 [152]. Hwang et al. showed that SCD1-dependent oleic acid provision by cancer-associated fibroblasts (CAFs) was essential in efficient xenograft lung carcinoma tumor formation [153]. Fritz et al. showed that advanced prostate carcinoma clinical biopsy samples display elevated SCD1 expression and that inhibition of SCD1 robustly inhibits prostate carcinoma xenograft growth [154].
Budhu et al. [155] and Ma et al. [156] found that elevated SCD1 is a negative prognostic in hepatocellular carcinoma. Peck et al. showed that SCD1 knockdown blocked prostate carcinoma xenograft growth and found that elevated SCD1 expression levels were a negative prognostic in breast carcinoma patients [141]. von Roemeling et al. showed that SCD1 is essential to anaplastic thyroid carcinoma (ATC) cell survival and elevated SCD1 levels are a negative prognostic in ATC patients [157]. Liu et al. found that elevated SCD1 is a robustly negative prognostic in pancreatic carcinoma, thymoma, melanoma, and renal clear cell carcinoma [158]. Noto et al. found that SCD1 is a strong negative prognostic in lung carcinoma and is necessary for the preservation of the stem cell-like status correlating with drug resistance [159,160]. Ran et al. showed that SCD1 expression was required for the epithelial-mesenchyme transition associated with in vivo metastasis and found that elevated SCD1 tumor expression was a robust negative clinical prognostic in colorectal carcinoma [161].
Li et al. showed that SCD1 is essential for ovarian carcinoma cell lines to form tumors in in vivo preclinical models [162]. Bansal et al. showed that hepatocellular carcinoma cell sensitivity to various chemotherapeutic agents was predicted by their levels of SCD1 expression; these resistance effects were attenuated by suppression of SCD1 expression or activity [163]. Luo et al. showed that SCD1 expression supported radiation resistance in esophageal squamous cell carcinoma [164]. Zhang et al. showed that SCD1 was essential to stem cell-like behavior and chemoresistance in gastric carcinoma [165]. Morais et al. [166] and Parik et al. [167] showed that glioblastoma aggressiveness and temozolomide resistance depended on SCD1 expression. Lien et al. showed that in vivo PDAC growth under low lipid dietary availability was strongly dependent on SCD1 [168]. Sun et al. showed that irinotecan resistance in colorectal carcinoma cells depended on SCD1 expression [169]. Li et al. showed that SCD1 ablation substantially inhibited endometrial carcinoma xenograft tumor growth [170].
Piao et al. generated cancer stem cell derivatives of a bladder carcinoma; these CSCs show substantially more efficient xenograft formation [171]. These authors then showed that SCD1 expression was prominently upregulated in these CSCs relative to the parental cell line and that SCD1 inhibition blocked in vitro migration and invasion of these CSCs. Finally, these authors found that high SCD1 expression was a strong negative prognostic for bladder carcinoma in clinical data sets.
Wang et al. found that high SCD1 expression was a strong negative prognostic factor in renal clear cell carcinoma [172].
While these data clearly indicate the pervasive involvement of SCD1 in support of carcinoma malignancy and treatment resistance, there are rare examples of the contrary case of SCD1 downregulation in treatment resistance [173,174]. It will ultimately be important to understand the mechanistic basis of these rare opposite response patterns.
The behavior of SCD1 in carcinomas and their treatment resistance commonly conform robustly to the involvement of peroxisomal fatty acid oxidation (Section 3.2) and mitochondrial redox reregulation (Section 3.1). Collectively, these data strongly support the LMRS hypothesis for treatment resistance. These insights indicate a path forward to effectively attacking this resistance, which, in turn, apparently drives the majority of carcinoma clinical lethality (below).

4. Conclusions and Practical Clinical Implications

The evidence above strongly suggests that the specialized LMRS lipid catabolic pathway is crucial to sustaining many or most cases of advanced carcinoma treatment resistance. These insights should allow an attack on the especially troublesome subclasses of carcinoma cells thought to be responsible for resistance; these include cells operationally classified as carcinoma stem cells, EMT-prone carcinoma cells, and/or carcinoma tumor initiating cells.
Most importantly, available evidence suggests that targeting the LMRS pathway may sensitize carcinoma cells to most or all of the diverse agents currently in wide use for cancer therapy. These range from radiation to chemotherapy/cytotoxic treatment, non-LMRS antimetabolic agents (CPI-613, for example), and immunotherapy. This perspective indicates a specific path toward improving many clinical outcomes, as follows: Table 1 summarizes selected preclinical studies that are especially useful for clinicians in exploring options they might wish to investigate.
One FDA-approved agent, thioridazine (TZ), attacks the peroxisomal fatty acid oxidase (Acox) step in the LMRS pathway (Figure 1). TZ targets Acox as a potent off-target effect [175,176]. TZ is relatively well tolerated, and clinicians have decades of experience with its chronic use in thousands of noncancer patients [177]. Thus, TZ represents an exceptionally promising opportunity to clinically test predictions of the LMRS hypothesis. Our preclinical experience with TZ in combination with the cancer mitochondrial TCA cycle inhibitor, CPI-613, supports this picture [121]. Likewise, Shen et al. [119] and Lasheras-Otero et al. [120] used TZ in their preclinical studies, supporting the generality of TZ use in carcinomas. Should TZ prove promising in initial clinical studies, the development of more robust Acox inhibitors would be warranted and likely practical.
As noted, inhibitor studies implicate acute MET kinase signaling in maintaining the LMRS pathway [121]. Consistent with this possibility, there is substantial evidence for MET upregulation in association with advanced carcinoma treatment resistance [178,179,180,181,182,183,184,185]. It will be important to assess the generality of MET inhibitors in apparently suppressing LMRS metabolism in carcinomas. The development of new MET inhibitors thrives [186,187,188,189].
Practical clinical assessment of attacks on other steps in the LMRS process is more remote than for TZ and CRZ. However, indications of success with the clinical application of TZ and/or CRZ would create the incentives to improve the targeting of the other steps in this process. The current status of these targeting opportunities is as follows:
There is evidence that the development of clinical inhibitors selective for the MCT2 transporter, essential to sustained peroxisomal oxidation, might be practical [189,190,191,192]. Such inhibitors are expected to potentially be as clinically effective as Acox inhibitors. Likewise, inhibitors of the CROT and CRAT transporters might be effective, though we currently lack any useful test cases for these.
While the involvement of SCD1 in carcinomas and, especially, in treatment resistance seems clear, practical clinical targeting of this enzyme will require careful additional exploration. Specifically, chronic SCD1 inhibition produces undesirable side effects, largely resulting from their compromising normal stem cell function [187]. However, it will be of value to explore whether acute SCD1 inhibition on the time scale of weeks might be sufficient to contribute to robust clinical anticarcinoma effects without unacceptable side effect toxicity. Small molecule SCD1 inhibitors are known, and the development of workable clinical versions is likely realistic [193,194].
Genetic ablation of NNT is relatively innocuous in the short term in humans [92,195], indicating that targeting this activity acutely in a therapeutic context should be well tolerated. Current experimental NNT inhibitors are clinically impractical [95]. Developing small molecule agents for clinical targeting of NNT is plausible in view of the detailed NNT structural picture now available [89].
Finally, of course, additional practical barriers to targeting the LMRS process might emerge. For example, some such approaches might counterproductively suppress immune attacks in carcinomas. Aggressive clinical and in vivo preclinical investigation of LMRS-related opportunities will be important in the near future.

Author Contributions

P.M.B. and Z.Z. contributed equally to this manuscript and to the experimental work from our group referred to herein. All authors have read and agreed to the published version of the manuscript.

Funding

Our experimental work in the area of this review [78,121] was funded by a FUSION Award from the Stony Brook School of Medicine, Stony Brook University, Stony Brook., NY, USA and by Cornerstone Pharmaceuticals Inc., Cranbury, NJ, USA.

Data Availability Statement

Not applicable.

Acknowledgments

We are grateful to our colleagues at Stony Brook University in the Biochemistry and Cell Biology Department and the Lipid Signaling and Metabolism in Cancer (LSMC) group for diverse helpful discussions.

Conflicts of Interest

P.M.B and Z.Z. invented and patented the CPI-613 drug family, through Stony Brook University. CPI-613 patents are currently licensed to Cornerstone Pharmaceuticals for clinical development.

References

  1. Dvorak, H.F. Tumors—Wounds that do not heal—Similarities between tumor stroma generation and wound-healing. N. Engl. J. Med. 1986, 315, 1650–1659. [Google Scholar] [CrossRef] [PubMed]
  2. Singer, A.J.; Clark, R.A.F. Mechanisms of disease—Cutaneous wound healing. N. Engl. J. Med. 1999, 341, 738–746. [Google Scholar] [CrossRef] [PubMed]
  3. Riss, J.; Khanna, C.; Koo, S.; Chandramouli, G.V.R.; Yang, H.H.; Hu, Y.; Kleiner, D.E.; Rosenwald, A.; Schaefer, C.F.; Ben-Sasson, S.A.; et al. Cancers as wounds that do not heal: Differences and similarities between renal regeneration/repair and renal cell carcinoma. Cancer Res. 2006, 66, 7216–7224. [Google Scholar] [CrossRef] [PubMed]
  4. Byun, J.S.; Gardner, K. Wounds that will not heal Pervasive cellular reprogramming in cancer. Am. J. Pathol. 2013, 182, 1055–1064. [Google Scholar] [CrossRef]
  5. Dvorak, H.F. Tumors: Wounds that do not heal-redux. Cancer Immunol. Res. 2015, 3, 1–11. [Google Scholar] [CrossRef] [PubMed]
  6. Dvorak, H.F. Tumors: Wounds that do not heal—A historical perspective with focus on the fundamental roles of increased vascular permeability and clotting. Semin. Thromb. Hemost. 2019, 45, 576–592. [Google Scholar] [CrossRef]
  7. Wong, A.Y.; Whited, J.L. Parallels between wound healing, epimorphic regeneration and solid tumors. Development 2020, 147, dev181636. [Google Scholar] [CrossRef]
  8. Deyell, M.; Garris, C.S.; Laughney, A.M. Cancer metastasis as a non-healing wound. Br. J. Cancer 2021, 124, 1491–1502. [Google Scholar] [CrossRef]
  9. Semenza, G.L. Hypoxia-inducible factors: Mediators of cancer progression and targets for cancer therapy. Trends Pharm. Sci. 2012, 33, 207–214. [Google Scholar] [CrossRef]
  10. Eales, K.L.; Hollinshead, K.E.R.; Tennant, D.A. Hypoxia and metabolic adaptation of cancer cells. Oncogenesis 2016, 5, e190. [Google Scholar] [CrossRef]
  11. Kamphorst, J.J.; Nofal, M.; Commisso, C.; Hackett, S.R.; Lu, W.Y.; Grabocka, E.; Vander Heiden, M.G.; Miller, G.; Drebin, J.A.; Bar-Sagi, D.; et al. Human Pancreatic Cancer Tumors Are Nutrient Poor and Tumor Cells Actively Scavenge Extracellular Protein. Cancer Res. 2015, 75, 544–553. [Google Scholar] [CrossRef] [PubMed]
  12. Stern, J.H.; Rutkowski, J.M.; Scherer, P.E. Adiponectin, leptin, and fatty acids in the maintenance of metabolic homeostasis through adipose tissue crosstalk. Cell Metab. 2016, 23, 770–784. [Google Scholar] [CrossRef] [PubMed]
  13. Snaebjornsson, M.T.; Janaki-Raman, S.; Schulze, A. Greasing the wheels of the cancer machine: The role of lipid metabolism in cancer. Cell Metab. 2020, 31, 62–76. [Google Scholar] [CrossRef] [PubMed]
  14. Martin-Perez, M.; Urdiroz-Urricelqui, U.; Bigas, C.; Benitah, S.A. The role of lipids in cancer progression and metastasis. Cell Metab. 2022, 34, 1675–1699. [Google Scholar] [CrossRef]
  15. Hatzivassiliou, G.; Zhao, F.P.; Bauer, D.E.; Andreadis, C.; Shaw, A.N.; Dhanak, D.; Hingorani, S.R.; Tuveson, D.A.; Thompson, C.B. ATP citrate lyase inhibition can suppress tumor cell growth. Cancer Cell 2005, 8, 311–321. [Google Scholar] [CrossRef]
  16. Currie, E.; Schulze, A.; Zechner, R.; Walther, T.C.; Farese, R.V. Cellular fatty acid metabolism and cancer. Cell Metab. 2013, 18, 153–161. [Google Scholar] [CrossRef]
  17. Jung, Y.Y.; Kim, H.M.; Koo, J.S. Expression of lipid metabolism-related proteins in metastatic breast cancer. PLoS ONE 2015, 10, e0137204. [Google Scholar] [CrossRef]
  18. Petan, T.; Jarc, E.; Jusovic, M. Lipid droplets in cancer: Guardians of fat in a stressful world. Molecules 2018, 23, 1941. [Google Scholar] [CrossRef]
  19. Biffi, G.; Tuveson, D.A. A FATal combination: Fibroblast-derived lipids and cancer-derived autotaxin promote pancreatic cancer growth. Cancer Disc. 2019, 9, 578–580. [Google Scholar] [CrossRef]
  20. Cao, Y.H. Adipocyte and lipid metabolism in cancer drug resistance. J. Clin. Investig. 2019, 129, 3006–3017. [Google Scholar] [CrossRef]
  21. Germain, N.; Dhayer, M.; Boileau, M.; Fovez, Q.; Kluza, J.; Marchetti, P. Lipid metabolism and resistance to anticancer treatment. Biology 2020, 9, 474. [Google Scholar] [CrossRef]
  22. Kopecka, J.; Trouillas, P.; Gasparovic, A.C.; Gazzano, E.; Assaraf, Y.G.; Riganti, C. Phospholipids and cholesterol: Inducers of cancer multidrug resistance and therapeutic targets. Drug Resist. Updates 2020, 49, 100670. [Google Scholar] [CrossRef] [PubMed]
  23. Biffi, G.; Tuveson, D.A. Diversity and biology of cancer-associated fibroblasts. Physiol. Rev. 2021, 101, 147–176. [Google Scholar] [CrossRef] [PubMed]
  24. Dlubek, J.; Rysz, J.; Jablonowski, Z.; Gluba-Brzozka, A.; Franczyk, B. The correlation between lipid metabolism disorders and prostate cancer. Curr. Med. Chem. 2021, 28, 2048–2061. [Google Scholar] [CrossRef] [PubMed]
  25. Fontaine, A.; Bellanger, D.; Guibon, R.; Bruyere, F.; Brisson, L.; Fromont, G. Lipophagy and prostate cancer: Association with disease aggressiveness and proximity to periprostatic adipose tissue. J. Path. 2021, 255, 166–176. [Google Scholar] [CrossRef] [PubMed]
  26. Hoy, A.J.; Nagarajan, S.R.; Butler, L.M. Tumour fatty acid metabolism in the context of therapy resistance and obesity. Nat. Rev. Cancer 2021, 21, 753–766. [Google Scholar] [CrossRef]
  27. Schiliro, C.; Firestein, B.L. Mechanisms of metabolic reprogramming in cancer cells supporting enhanced growth and proliferation. Cells 2021, 10, 1056. [Google Scholar] [CrossRef]
  28. An, D.D.; Zhai, D.Y.; Wan, C.; Yang, K.Y. The role of lipid metabolism in cancer radioresistance. Clin. Transl. Oncol. 2023, 25, 2332–2349. [Google Scholar] [CrossRef]
  29. Hoang-Minh, L.B.; Siebzehnrubl, F.A.; Yang, C.L.; Suzuki-Hatano, S.; Dajac, K.; Loche, T.; Andrews, N.; Massari, M.S.; Patel, J.; Amin, K.; et al. Infiltrative and drug-resistant slow-cycling cells support metabolic heterogeneity in glioblastoma. EMBO J. 2018, 37, e98772. [Google Scholar] [CrossRef]
  30. Aloia, A.; Mullhaupt, D.; Chabbert, C.D.; Eberhart, T.; Fluckiger-Mangual, S.; Vukolic, A.; Eichhoff, O.; Irmisch, A.; Alexander, L.T.; Scibona, E. A fatty acid oxidation-dependent metabolic shift regulates the adaptation of BRAF-mutated melanoma to MAPK inhibitors. J. Clin. Investig. 2019, 25, 6852–6867. [Google Scholar] [CrossRef]
  31. Beloribi-Djefaflia, S.; Vasseur, S.; Guillaumond, F. Lipid metabolic reprogramming in cancer cells. Oncogenesis 2016, 5, e189. [Google Scholar] [CrossRef]
  32. Sunami, Y.; Rebelo, A.; Kleeff, J. Lipid metabolism and lipid droplets in pancreatic cancer and stellate cells. Cancers 2018, 10, 3. [Google Scholar] [CrossRef]
  33. Benzarti, M.; Delbrouck, C.; Neises, L.; Kiweler, N.; Meiser, J. Metabolic potential of cancer cells in context of the metastatic cascade. Cells 2020, 9, 2035. [Google Scholar] [CrossRef]
  34. Azam, A.; Sounni, N.E. Lipid metabolism heterogeneity and crosstalk with mitochondria functions drive breast cancer progression and drug resistance. Cancers 2022, 14, 6267. [Google Scholar] [CrossRef] [PubMed]
  35. Miao, X.W.; Wang, B.L.; Chen, K.L.; Ding, R.; Wu, J.C.; Pan, Y.; Ji, P.; Ye, B.; Xiang, M.L. Perspectives of lipid metabolism reprogramming in head and neck squamous cell carcinoma: An overview. Front. Oncol. 2022, 12, 1008361. [Google Scholar] [CrossRef] [PubMed]
  36. Yang, R.X.; Yi, M.; Xiang, B. Novel insights on lipid metabolism alterations in drug resistance in cancer. Front. Cell Dev. Biol. 2022, 10, 875318. [Google Scholar] [CrossRef] [PubMed]
  37. Bensaad, K.; Favaro, E.; Lewis, C.A.; Peck, B.; Lord, S.; Collins, J.M.; Pinnick, K.E.; Wigfield, S.; Buffa, F.M.; Li, J.-L. Fatty acid uptake and lipid storage induced by HIF-1 alpha contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep. 2014, 9, 349–365. [Google Scholar] [CrossRef]
  38. Rohwer, N.; Cramer, T. Hypoxia-mediated drug resistance: Novel insights on the functional interaction of HIFs and cell death pathways. Drug Resist. Updates 2011, 14, 191–201. [Google Scholar] [CrossRef]
  39. Munir, R.; Lisec, J.; Swinnen, J.V.; Zaidi, N. Lipid metabolism in cancer cells under metabolic stress. Br. J. Cancer 2019, 120, 1090–1098. [Google Scholar] [CrossRef]
  40. Xie, H.; Simon, M.C. Oxygen availability and metabolic reprogramming in cancer. J. Biol. Chem. 2017, 292, 16825–16832. [Google Scholar] [CrossRef]
  41. Zhang, X.D.; Saarinen, A.M.; Hitosugi, T.; Wang, Z.H.; Wang, L.G.; Ho, T.H.; Liu, J. Inhibition of intracellular lipolysis promotes human cancer cell adaptation to hypoxia. eLife 2017, 6, e31132. [Google Scholar] [CrossRef] [PubMed]
  42. Castelli, S.; Ciccarone, F.; Tavian, D.; Ciriolo, M.R. ROS-dependent HIF1 alpha activation under forced lipid catabolism entails glycolysis and mitophagy as mediators of higher proliferation rate in cervical cancer cells. J. Exp. Clin. Caner Res. 2021, 40, 94. [Google Scholar] [CrossRef]
  43. Chun, Y.; Kim, J. AMPK-mTOR Signaling and cellular adaptations in hypoxia. Int. J. Mol. Sci. 2021, 22, 9765. [Google Scholar] [CrossRef] [PubMed]
  44. Infantino, V.; Santarsiero, A.; Convertini, P.; Todisco, S.; Iacobazzi, V. Cancer cell metabolism in hypoxia: Role of HIF-1 as key regulator and therapeutic target. Int. J. Mol. Sci. 2021, 22, 5703. [Google Scholar] [CrossRef] [PubMed]
  45. Seo, J.; Yun, J.E.; Kim, S.J.; Chun, Y.S. Lipid metabolic reprogramming by hypoxia-inducible factor-1 in the hypoxic tumour microenvironment. Pflug. Arch. Eur. J. Physiol. 2022, 474, 591–601. [Google Scholar] [CrossRef] [PubMed]
  46. Salavaty, A.; Azadian, E.; Naik, S.H.; Currie, P.D. Clonal selection parallels between normal and cancer tissues. Trend Genet. 2023, 39, 358–380. [Google Scholar] [CrossRef]
  47. Vasan, K.; Werner, M.; Chandel, N.S. Mitochondrial metabolism as a target for cancer therapy. Cell Metab. 2020, 32, 341–352. [Google Scholar] [CrossRef]
  48. Pavlova, N.N.; Zhu, J.J.; Thompson, C.B. The hallmarks of cancer metabolism: Still emerging. Cell Metab. 2022, 34, 355–377. [Google Scholar] [CrossRef]
  49. Lasorda, F.; di Meo, N.A.; Rutigliano, M.; Ferro, M.; Terracciano, D.; Tataru, O.S.; Battaglia, M.; Ditonno, P.; Lucarelli, G. Emerging hallmarks of metabolic reprogramming in prostate cancer. Int. J. Mol. Sci. 2023, 24, 910. [Google Scholar] [CrossRef]
  50. Li, Y.J.; Zhang, C.Y.; Martincuks, A.; Herrmann, A.; Yu, H. STAT proteins in cancer: Orchestration of metabolism. Nat. Rev. Cancer 2023, 23, 115–134. [Google Scholar] [CrossRef]
  51. Tadros, S.; Shukla, S.K.; King, R.J.; Gunda, V.; Vernucci, E.; Abrego, J.; Chaika, N.V.; Yu, F.; Lazenby, A.J.; Berim, L.; et al. De novo lipid synthesis facilitates gemcitabine resistance through endoplasmic reticulum stress in pancreatic cancer. Cancer Res. 2017, 77, 5503–5517. [Google Scholar] [CrossRef] [PubMed]
  52. Menendez, J.A.; Lupu, R. Fatty acid synthase: A druggable driver of breast cancer brain metastasis. Expert Opin. Ther. Targets 2022, 26, 427–444. [Google Scholar] [CrossRef] [PubMed]
  53. Zhou, Y.; Tao, J.Y.; Calvisi, D.F.; Chen, X. Role of lipogenesis rewiring in hepatocellular carcinoma. Sem. Liver Dis. 2022, 42, 77–86. [Google Scholar] [CrossRef] [PubMed]
  54. Kawaguchi, K.; Kinameri, A.; Suzuki, S.; Senga, S.; Ke, Y.Q.; Fujii, H. The cancer-promoting gene fatty acid-binding protein 5 (FABP5) is epigenetically regulated during human prostate carcinogenesis. Biochem. J. 2016, 473, 449–461. [Google Scholar] [CrossRef] [PubMed]
  55. Pascual, G.; Avgustinova, A.; Mejetta, S.; Martin, M.; Castellanos, A.; Attolini, C.S.O.; Berenguer, A.; Prats, N.; Toll, A.; Hueto, J.A.; et al. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature 2017, 541, 41–49. [Google Scholar] [CrossRef]
  56. Cheng, C.M.; Geng, F.; Cheng, X.; Guo, D.L. Lipid metabolism reprogramming and its potential targets in cancer. Cancer Commun. 2018, 38, 27. [Google Scholar] [CrossRef]
  57. Su, F.; Ahn, S.; Saha, A.; DiGiovanni, J.; Kolonin, M.G. Adipose stromal cell targeting suppresses prostate cancer epithelial-mesenchymal transition and chemoresistance. Oncogene 2019, 38, 1979–1988. [Google Scholar] [CrossRef]
  58. Wang, J.C.; Li, Y.S. CD36 tango in cancer: Signaling pathways and functions. Theranostics 2019, 9, 4893–4908. [Google Scholar] [CrossRef]
  59. Nieman, K.M.; Kenny, H.A.; Penicka, C.V.; Ladanyi, A.; Buell-Gutbrod, R.; Zillhardt, M.R.; Romero, I.L.; Carey, M.S.; Mills, G.B.; Hotamisligil, G.S.; et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat. Med. 2011, 17, 1498-U1207. [Google Scholar] [CrossRef]
  60. Singh, S.R.; Zeng, X.K.; Zhao, J.S.; Liu, Y.; Hou, G.; Liu, H.H.; Hou, S.X. The lipolysis pathway sustains normal and transformed stem cells in adult Drosophila. Nature 2016, 538, 109–115. [Google Scholar] [CrossRef]
  61. Okumura, T.; Ohuchida, K.; Sada, M.; Abe, T.; Endo, S.; Koikawa, K.; Iwamoto, C.; Miura, D.; Mizuuchi, Y.; Moriyama, T.; et al. Extra-pancreatic invasion induces lipolytic and fibrotic changes in the adipose microenvironment, with released fatty acids enhancing the invasiveness of pancreatic cancer cells. Oncotarget 2017, 8, 18280–18295. [Google Scholar] [CrossRef] [PubMed]
  62. Wang, Y.Y.; Attane, C.; Milhas, D.; Dirat, B.; Dauvillier, S.; Guerard, A.; Gilhodes, J.; Lazar, I.; Alet, N.; Laurent, V.; et al. Mammary adipocytes stimulate breast cancer invasion through metabolic remodeling of tumor cells. JCI Insight 2017, 2, e87489. [Google Scholar] [CrossRef] [PubMed]
  63. Ladanyi, A.; Mukherjee, A.; Kenny, H.A.; Johnson, A.; Mitra, A.K.; Sundaresan, S.; Nieman, K.M.; Pascual, G.; Benitah, A.S.; Montag, A.; et al. Adipocyte-induced CD36 expression drives ovarian cancer progression and metastasis. Oncogene 2018, 37, 2285–2301. [Google Scholar] [CrossRef] [PubMed]
  64. Auciello, F.R.; Bulusu, V.; Oon, C.; Tat-Mulder, J.; Berry, M.; Bhattacharyya, S.; Tumanov, S.; Allen-Petersen, B.L.; Link, J.; Kendsersky, N.D.; et al. A stromal lysolipid-autotaxin signaling axis promotes pancreatic tumor progression. Cell Discov. 2019, 9, 617–627. [Google Scholar] [CrossRef] [PubMed]
  65. Gong, J.; Lin, Y.Y.; Zhang, H.Q.; Liu, C.Q.; Cheng, Z.; Yang, X.W.; Zhang, J.; Xiao, Y.; Sang, N.; Qian, X.; et al. Reprogramming of lipid metabolism in cancer-associated fibroblasts potentiates migration of colorectal cancer cells. Cell Death Dis. 2020, 11, 267. [Google Scholar] [CrossRef]
  66. Kay, E.J.; Zanivan, S. Metabolic pathways fuelling protumourigenic cancer-associated fibroblast functions. Curr. Opin. Syst. Biol. 2021, 28, 28. [Google Scholar] [CrossRef]
  67. Sugai, M.; Yanagawa, N.; Shikanai, S.; Hashimoto, M.; Saikawa, H.; Osakabe, M.; Saito, H.; Maemondo, M.; Sugai, T. Correlation of tumor microenvironment-related markers with clinical outcomes in patients with squamous cell carcinoma of the lung. Transl. Lung Cancer Res. 2022, 11, 975–985. [Google Scholar] [CrossRef]
  68. Andreucci, E.; Fioretto, B.S.; Rosa, I.; Matucci-Cerinic, M.; Biagioni, A.; Romano, E.; Calorini, L.; Manetti, M. Extracellular Lactic Acidosis of the Tumor Microenvironment Drives Adipocyte-to-Myofibroblast Transition Fueling the Generation of Cancer-Associated Fibroblasts. Cells 2023, 12, 939. [Google Scholar] [CrossRef]
  69. Hamabe-Horiike, T.; Harada, S.I.; Yoshida, K.; Kinoshita, J.; Yamaguchi, T.; Fushida, S. Adipocytes contribute to tumor progression and invasion of peritoneal metastasis by interacting with gastric cancer cells as cancer associated fibroblasts. Cancer Rep. 2023, 6, e1647. [Google Scholar] [CrossRef]
  70. Sato, S.; Hiruma, T.; Koizumi, M.; Yoshihara, M.; Nakamura, Y.; Tadokoro, H.; Motomatsu, S.; Yamanaka, T.; Washimi, K.; Okubo, Y.; et al. Bone marrow adipocytes induce cancer-associated fibroblasts and immune evasion, enhancing invasion and drug resistance. Cancer Sci. 2023, 114, 2674–2688. [Google Scholar] [CrossRef]
  71. Nieman, K.M.; Romero, I.L.; Van Houten, B.; Lengyel, E. Adipose tissue and adipocytes support tumorigenesis and metastasis. BBA Mol. Cell Biol. Lipids 2013, 1831, 1533–1541. [Google Scholar] [CrossRef] [PubMed]
  72. Lengyel, E.; Makowski, L.; DiGiovanni, J.; Kolonin, M.G. Cancer as a matter of fat: The crosstalk between adipose tissue and tumors. Trends Cancer 2018, 4, 374–384. [Google Scholar] [CrossRef] [PubMed]
  73. Chen, X.M.; Song, E.W. Turning foes to friends: Targeting cancer-associated fibroblasts. Nat. Rev. Drug Discov. 2019, 18, 99–115. [Google Scholar] [CrossRef] [PubMed]
  74. Sacca, P.A.; Calvo, J.C. Periprostatic adipose tissue microenvironment: Metabolic and hormonal pathways during prostate cancer progression. Front. Endocrinol. 2022, 13, 863027. [Google Scholar] [CrossRef]
  75. Sarkar, M.; Nguyen, T.; Gundre, E.; Ogunlusi, O.; El-Sobky, M.; Giri, B.; Sarkar, T.R. Cancer-associated fibroblasts: The chief architect in the tumor microenvironment. Front. Cell Dev. Biol. 2023, 11, 1089068. [Google Scholar] [CrossRef]
  76. Sherman, M.H.; di Magliano, M.P. Cancer-associated fibroblasts: Lessons from pancreatic cancer. Annu. Rev. Cancer Biol. 2023, 7, 43–55. [Google Scholar] [CrossRef]
  77. Mailloux, R.J. Mitochondrial antioxidants and the maintenance of cellular hydrogen peroxide levels. Oxidative Med. Cell. Longev. 2018, 2018, 7857251. [Google Scholar] [CrossRef]
  78. Stuart, S.D.; Schauble, A.; Gupta, S.; Kennedy, A.D.; Keppler, B.R.; Bingham, P.M.; Zachar, Z. A strategically designed small molecule attacks alpha-ketoglutarate dehydrogenase in tumor cells through a redox process. Cancer Metab. 2014, 2, 4. [Google Scholar] [CrossRef]
  79. McLain, A.L.; Szweda, P.A.; Szweda, L.I. alpha-Ketoglutarate dehydrogenase: A mitochondrial redox sensor. Free Rad. Res. 2011, 45, 29–36. [Google Scholar] [CrossRef]
  80. Solmonson, A.; DeBerardinis, R.J. Lipoic acid metabolism and mitochondrial redox regulation. J. Biol. Chem. 2018, 293, 7522–7530. [Google Scholar] [CrossRef]
  81. Mailloux, R.J.; Grayson, C.; Koufos, O. Regulation of mitochondrial hydrogen peroxide availability by protein S-glutathionylation. Cells 2023, 12, 107. [Google Scholar] [CrossRef]
  82. Tonazzi, A.; Giangregorio, N.; Console, L.; Palmieri, F.; Indiveri, C. The mitochondrial carnitine acyl-carnitine carrier (SLC25A20): Molecular mechanisms of transport, role in redox sensing and interaction with drugs. Biomolecules 2021, 11, 521. [Google Scholar] [CrossRef] [PubMed]
  83. Kisty, E.A.; Saart, E.C.; Weerapana, E. Identifying redox-sensitive cysteine residues in mitochondria. Antioxidants 2023, 12, 992. [Google Scholar] [CrossRef] [PubMed]
  84. Lushchak, O.V.; Piroddi, M.; Galli, F.; Lushchak, V.I. Aconitase post-translational modification as a key in linkage between Krebs cycle, iron homeostasis, redox signaling, and metabolism of reactive oxygen species. Redox Rep. 2014, 19, 8–15. [Google Scholar] [CrossRef] [PubMed]
  85. Wong, H.S.; Dighe, P.A.; Mezera, V.; Monternier, P.A.; Brand, M.D. Production of superoxide and hydrogen peroxide from specific mitochondrial sites under different bioenergetic conditions. J. Biol. Chem. 2017, 292, 16804–16809. [Google Scholar] [CrossRef] [PubMed]
  86. Castelli, S.; De Falco, P.; Ciccarone, F.; Desideri, E.; Ciriolo, M.R. Lipid catabolism and ROS in cancer: A bidirectional liaison. Cancers 2021, 13, 5484. [Google Scholar] [CrossRef]
  87. Castelli, S.; Ciccarone, F.; De Falco, P.; Ciriolo, M.R. Adaptive antioxidant response to mitochondrial fatty acid oxidation determines the proliferative outcome of cancer cells. Cancer Lett. 2023, 554, 216010. [Google Scholar] [CrossRef]
  88. Ying, M.F.; Hu, X. Tracing the electron flow in redox metabolism: The appropriate distribution of electrons is essential to maintain redox balance in cancer cells. Semin. Cancer Biol. 2022, 87, 32–47. [Google Scholar] [CrossRef]
  89. Kampjut, D.; Sazanov, L.A. Structure and mechanism of mitochondrial proton-translocating transhydrogenase. Nature 2019, 573, 291–295. [Google Scholar] [CrossRef]
  90. Jackson, J.B. A review of the binding-change mechanism for proton-translocating transhydrogenase. BBA Bioenerg. 2012, 1817, 1839–1846. [Google Scholar] [CrossRef]
  91. Nesci, S.; Trombetti, F.; Pagliarani, A. Nicotinamide nucleotide transhydrogenase as a sensor of mitochondrial biology. Trends Cell Biol. 2020, 30, 1–3. [Google Scholar] [CrossRef]
  92. Francisco, A.; Figueira, T.R.; Castilho, R.F. Mitochondrial NAD(P)(+) transhydrogenase: From molecular features to physiology and disease. Antioxid. Redox Signal. 2022, 36, 864–884. [Google Scholar] [CrossRef] [PubMed]
  93. Rydstrom, J. Site-specific inhibitors of mitochondrial nicotinamide-nucleotide transhydrogenase. Eur. J. Biochem. 1972, 31, 496–504. [Google Scholar] [CrossRef] [PubMed]
  94. Bicego, R.; Francisco, A.; Ruas, J.S.; Siqueira-Santos, E.S.; Castilho, R.F. Undesirable effects of chemical inhibitors of NAD(P)(+) transhydrogenase on mitochondrial respiratory function. Arch. Biochem. Biophys. 2020, 692, 692. [Google Scholar] [CrossRef] [PubMed]
  95. Rydstrom, J. Mitochondrial NADPH, transhydrogenase and disease. BBA Bioenerg. 2006, 1757, 721–726. [Google Scholar] [CrossRef]
  96. Han, Y.; Zhang, Y.Y.; Pan, Y.Q.; Zheng, X.J.; Liao, K.; Mo, H.Y.; Sheng, H.; Wu, Q.-N.; Liu, Z.-X.; Zeng, Z.-L.; et al. IL-10-associated NNT acetylation orchestrates iron-sulfur cluster maintenance and cancer immunotherapy resistance. Mol. Cell 2023, 83, 1887–1895. [Google Scholar] [CrossRef]
  97. Chortis, V.; Taylor, A.E.; Doig, C.L.; Walsh, M.D.; Meimaridou, E.; Jenkinson, C.; Rodriguez-Blanco, G.; Ronchi, C.L.; Jafri, A.; Metherell, L.A.; et al. Nicotinamide nucleotide transhydrogenase as a novel treatment target in adrenocortical carcinoma. Endocrinology 2018, 159, 2836–2849. [Google Scholar] [CrossRef]
  98. Gameiro, P.A.; Laviolette, L.A.; Kelleher, J.K.; Iliopoulos, O.; Stephanopoulos, G. Cofactor balance by nicotinamide nucleotide transhydrogenase (NNT) coordinates reductive carboxylation and glucose catabolism in the tricarboxylic acid (TCA) cycle. J. Biol. Chem. 2013, 288, 12967–12977. [Google Scholar] [CrossRef]
  99. Li, S.; Zhuang, Z.N.; Wu, T.; Lin, J.C.; Liu, Z.X.; Zhou, L.F.; Dai, T.; Lu, L.; Ju, H.Q. Nicotinamide nucleotide transhydrogenase-mediated redox homeostasis promotes tumor growth and metastasis in gastric cancer. Redox Biol. 2018, 18, 246–255. [Google Scholar] [CrossRef]
  100. Ward, N.P.; Kang, Y.P.; Falzone, A.; Boyle, T.A.; DeNicola, G.M. Nicotinamide nucleotide transhydrogenase regulates mitochondrial metabolism in NSCLC through maintenance of Fe-S protein function. J. Exp. Med. 2020, 217, e20191689. [Google Scholar] [CrossRef]
  101. Mullen, A.R.; Hu, Z.; Shi, X.; Jiang, L.; Boroughs, L.K.; Kovacs, Z.; Boriack, R.; Rakheja, D.; Sullivan, L.B.; Linehan, W.M.; et al. Oxidation of alpha-ketoglutarate is required for reductive carboxylation in cancer cells with mitochondrial defects. Cell Rep. 2014, 7, 1679–1690. [Google Scholar] [CrossRef] [PubMed]
  102. Duan, Z.J.; Song, Y.; Zou, X.J.; Liu, S.S.; Zhang, W.L.; Liu, L. Nicotinamide nucleotide transhydrogenase acts as a new prognosis biomarker in hepatocellular carcinoma. Int. J. Clin. Exp. Pathol. 2020, 13, 972–978. [Google Scholar] [PubMed]
  103. Lin, X.J.; Wu, W.Z.; Ying, Y.K.; Luo, J.; Xu, X.H.; Zheng, L.X.; Wu, W.; Yang, S.; Zhao, S.K. MicroRNA-31: A pivotal oncogenic factor in oral squamous cell carcinoma. Cell Death Discov. 2022, 8, 140. [Google Scholar] [CrossRef] [PubMed]
  104. Wanders, R.J.A.; Waterham, H.R.; Ferdinandusse, S. Metabolic Interplay between peroxisomes and other subcellular organelles including mitochondria and the endoplasmic reticulum. Front. Cell Dev. Biol. 2016, 3, 83. [Google Scholar] [CrossRef] [PubMed]
  105. Fransen, M.; Lismont, C.; Walton, P. The peroxisome-mitochondria connection: How and why? Int. J. Mol. Sci. 2017, 18, 1126. [Google Scholar] [CrossRef]
  106. Henne, W.M.; Reese, M.L.; Goodman, J.M. The assembly of lipid droplets and their roles in challenged cells. EMBO J. 2018, 37, e98947. [Google Scholar] [CrossRef]
  107. Klemm, R.W. Getting in touch is an important step: Control of metabolism at organelle contact sites. Sage J. 2021, 4, 2515256421993708. [Google Scholar] [CrossRef]
  108. Renne, M.F.; Hariri, H. Lipid droplet-organelle contact sites as hubs for fatty acid metabolism, trafficking, and metabolic channeling. Front. Cell Dev. Biol. 2021, 9, 726261. [Google Scholar] [CrossRef]
  109. Kim, J.A. Peroxisome metabolism in cancer. Cells 2020, 9, 1692. [Google Scholar] [CrossRef]
  110. Dahabieh, M.S.; Di Pietro, E.; Jangal, M.; Goncalves, C.; Witcher, M.; Braverman, N.E.; del Rincon, S.V. Peroxisomes and cancer: The role of a metabolic specialist in a disease of aberrant metabolism. BBA Rev. Cancer 2018, 1870, 103–121. [Google Scholar] [CrossRef]
  111. Ravi, A.; Palamiuc, L.; Emerling, B.M. Crucial players for inter-organelle communication: PI5P4Ks and their lipid product PI-4,5-P-2 come to the surface. Front. Cell Dev. Biol. 2022, 9, 791758. [Google Scholar] [CrossRef] [PubMed]
  112. Schlaepfer, I.R.; Joshi, M. CPT1A-mediated fat oxidation, mechanisms, and therapeutic potential. Endocrinology 2020, 161, bqz046. [Google Scholar] [CrossRef] [PubMed]
  113. Waterham, H.R.; Ferdinandusse, S.; Wanders, R.J.A. Human disorders of peroxisome metabolism and biogenesis. BBA Mol. Cell Res. 2016, 1863, 922–933. [Google Scholar] [CrossRef] [PubMed]
  114. Cai, M.J.; Sun, X.; Wang, W.C.; Lian, Z.S.; Wu, P.; Han, S.X.; Chen, H.; Zhang, P.M. Disruption of peroxisome function leads to metabolic stress, mTOR inhibition, and lethality in liver cancer cells. Caner Lett. 2018, 421, 82–93. [Google Scholar] [CrossRef]
  115. Koukourakis, M.I.; Giatromanolaki, A.; Harris, A.L.; Sivridis, E. Comparison of metabolic pathways between cancer cells and stromal cells in colorectal carcinomas: A metabolic survival role for tumor-associated stroma. Cancer Res. 2006, 66, 632–637. [Google Scholar] [CrossRef]
  116. Valenca, I.; Pertega-Gomes, N.; Vizcaino, J.R.; Henrique, R.M.; Lopes, C.; Baltazar, F.; Ribeiro, D. Localization of MCT2 at peroxisomes is associated with malignant transformation in prostate cancer. J. Cell. Mol. Med. 2015, 19, 723–733. [Google Scholar] [CrossRef]
  117. Valenca, I.; Ferreira, A.R.; Correia, M.; Kuhl, S.; van Roermund, C.; Waterham, H.R.; Maximo, V.; Islinger, M.; Ribeiro, D. Prostate cancer proliferation is affected by the subcellular localization of MCT2 and accompanied by significant peroxisomal alterations. Cancers 2020, 12, 3152. [Google Scholar] [CrossRef]
  118. Kane, D.A. Lactate oxidation at the mitochondria: A lactate-malate-aspartate shuttle at work. Front. Neurosci. 2014, 8, 366. [Google Scholar] [CrossRef]
  119. Shen, S.S.; Faouzi, S.; Souquere, S.; Roy, S.; Routier, E.; Libenciuc, C.; Andre, F.; Pierron, G.; Scoazec, J.-Y.; Robert, C. Melanoma persister cells are tolerant to BRAF/MEK inhibitors via ACOX1-mediated fatty acid oxidation. Cell Rep. 2020, 33, 108421. [Google Scholar] [CrossRef]
  120. Lasheras-Otero, I.; Feliu, I.; Maillo, A.; Moreno, H.; Redondo-Munoz, M.; Aldaz, P.; Bocanegra, A.; Olias-Arjona, A.; Lecanda, F.; Fernandez-Irigoyen, J.; et al. The regulators of peroxisomal acyl-carnitine shuttle CROT and CRAT promote metastasis in melanoma. J. Investig. Dermatol. 2023, 143, 305–318. [Google Scholar] [CrossRef]
  121. Guardado Rivas, M.O.; Stuart, S.D.; Thach, D.; Dahan, M.; Shorr, R.; Zachar, Z.; Bingham, P.M. Evidence for a novel, effective approach to targeting carcinoma catabolism exploiting the first-in-class, anti-cancer mitochondrial drug, CPI-613. PLoS ONE 2022, 17, e0269620. [Google Scholar] [CrossRef] [PubMed]
  122. Lauer, C.; Volkl, A.; Riedl, S.; Fahimi, H.D.; Beier, K. Impairment of peroxisomal biogenesis in human colon carcinoma. Carcinogenesis 1999, 20, 985–989. [Google Scholar] [CrossRef] [PubMed]
  123. Frederiks, W.M.; Bosch, K.S.; Hoeben, K.A.; van Marle, J.; Langbein, S. Renal cell carcinoma and oxidative stress: The lack of peroxisomes. Acta Histochem. 2010, 112, 364–371. [Google Scholar] [CrossRef] [PubMed]
  124. Dahabieh, M.S.; Ha, Z.Y.; Di Pietro, E.; Nichol, J.N.; Bolt, A.M.; Goncalves, C.; Dupere-Richer, D.; Pettersson, F.; Mann, K.K.; Braverman, N.E.; et al. Peroxisomes protect lymphoma cells from HDAC inhibitor-mediated apoptosis. Cell Death Differ. 2017, 24, 1912–1924. [Google Scholar] [CrossRef] [PubMed]
  125. Dahabieh, M.S.; Huang, F.; Goncalves, C.; Gonzalez, R.E.F.; Prabhu, S.; Bolt, A.; Di Pietro, E.; Khoury, E.; Heath, J.; Xu, Z.Y.; et al. Silencing PEX26 as an unconventional mode to kill drug-resistant cancer cells and forestall drug resistance. Autophagy 2022, 18, 540–558. [Google Scholar] [CrossRef]
  126. Zheng, F.M.; Chen, W.B.; Qin, T.; Lv, L.N.; Feng, B.; Lu, Y.L.; Li, Z.-Q.; Wang, X.-C.; Tao, L.-J.; Li, H.-W.; et al. ACOX1 destabilizes p73 to suppress intrinsic apoptosis pathway and regulates sensitivity to doxorubicin in lymphoma cells. BMB Rep. 2019, 52, 566–571. [Google Scholar] [CrossRef]
  127. Kim, S.; Lee, Y.; Koo, J.S. Differential expression of lipid metabolism-related proteins in different breast cancer subtypes. PLoS ONE 2015, 10, e0119473. [Google Scholar] [CrossRef]
  128. Yu, G.Y.; Cheng, C.J.; Lin, S.C.; Lee, Y.C.; Frigo, D.E.; Yu-Lee, L.Y.; Gallick, G.E.; Titus, M.A.; Nutt, L.K.; Lin, S.H. Organelle-derived acetyl-CoA promotes prostate cancer cell survival, migration, and metastasis via activation of calmodulin kinase II. Cancer Res. 2018, 78, 2490–2502. [Google Scholar] [CrossRef]
  129. Kuna, R.S.; Kumar, A.; Wessendorf-Rodriguez, K.A.; Galvez, H.; Green, C.R.; McGregor, G.H.; Cordes, T.; Shaw, R.J.; Svensson, R.U.; Metallo, C.M. Inter-organelle cross-talk supports acetyl-coenzyme A homeostasis and lipogenesis under metabolic stress. Sci. Adv. 2023, 9, eadf0138. [Google Scholar] [CrossRef]
  130. Tamatani, T.; Hattori, K.; Nakashiro, K.I.; Hayashi, Y.; Wu, S.Q.; Klumpp, D.; Reddy, J.K.; Oyasu, R. Neoplastic conversion of human urothelial cells in vitro by overexpression of H2O2-generating peroxisomal fatty acyl CoA oxidase. Int. J. Oncol. 1999, 15, 743–749. [Google Scholar] [CrossRef]
  131. Okamoto, M.; Reddy, J.K.; Oyasu, R. Tumorigenic conversion of a non-tumorigenic rat urothelial cell line by overexpression of H2O2-generating peroxisomal fatty acyl-CoA oxidase. Int. J. Cancer 1997, 70, 716–721. [Google Scholar] [CrossRef]
  132. Sellin, J.; Wingen, C.; Gosejacob, D.; Senyilmaz, D.; Hanschke, L.; Buttner, S.; Meyer, K.; Bano, D.; Nicotera, P.; Teleman, A.A.; et al. Dietary rescue of lipotoxicity-induced mitochondrial damage in Peroxin19 mutants. PLoS Biol. 2018, 16, e2004893. [Google Scholar] [CrossRef]
  133. Ben-David, U.; Gan, Q.F.; Golan-Lev, T.; Arora, P.; Yanuka, O.; Oren, Y.S.; Leikin-Frenkel, A.; Graf, M.; Garippa, R.; Boehringer, M.; et al. Selective elimination of human pluripotent stem cells by an oleate synthesis inhibitor discovered in a high-throughput screen. Cell Stem Cell 2013, 12, 167–179. [Google Scholar] [CrossRef]
  134. Mohammadzadeh, F.H.V.; Alihemmati, A.; Shaaker, M.; Mosayyeb, G.; Darabi, M.; Mehdizadeh, A. The role of stearoyl-coenzyme A desaturase 1 in liver development, function, and pathogenesis. J. Ren. Hepatic Disord. 2019, 3, 15–22. [Google Scholar] [CrossRef]
  135. Xue, Y.Q.; Lin, L.Y.; Li, Q.; Liu, K.L.; Hu, M.Y.; Ye, J.Y.; Cao, J.; Zhai, J.; Zheng, F.; Wang, Y.; et al. SCD1 sustains homeostasis of bulge niche via maintaining hemidesmosomes in basal keratinocytes. Adv. Sci. 2023, 10, 2201949. [Google Scholar] [CrossRef] [PubMed]
  136. Aljohani, A.M.; Syed, D.N.; Ntambi, J.M. Insights into stearoyl-coa desaturase-1 regulation of systemic metabolism. Trends Endocrinol. Metab. 2017, 28, 831–842. [Google Scholar] [CrossRef]
  137. Paton, C.M.; Ntambi, J.M. Biochemical and physiological function of stearoyl-CoA desaturase. Am. J. Physiol.-Endocrinol. Metab. 2009, 297, E28–E37. [Google Scholar] [CrossRef]
  138. Igal, R.A. Stearoyl CoA desaturase-1: New insights into a central regulator of cancer metabolism. BBA Mol. Cell Biol. Lipids 2016, 1861, 1865–1880. [Google Scholar] [CrossRef]
  139. Ascenzi, F.; De Vitis, C.; Maugeri-Sacca, M.; Napoli, C.; Ciliberto, G.; Mancini, R. SCD1, autophagy and cancer: Implications for therapy. J. Exp. Clin. Cancer Res. 2021, 40, 265. [Google Scholar] [CrossRef]
  140. Young, R.M.; Ackerman, D.; Quinn, Z.L.; Mancuso, A.; Gruber, M.; Liu, L.P.; Giannoukos, D.N.; Bobrovnikova-Marjon, E.; Diehl, J.A.; Keith, B.; et al. Dysregulated mTORC1 renders cells critically dependent on desaturated lipids for survival under tumor-like stress. Genes Dev. 2013, 27, 1115–1131. [Google Scholar] [CrossRef]
  141. Peck, B.; Schug, Z.T.; Zhang, Q.; Dankworth, B.; Jones, D.T.; Smethurst, E.; Patel, R.; Mason, S.; Jiang, M.; Saunders, R.; et al. Inhibition of fatty acid desaturation is detrimental to cancer cell survival in metabolically compromised environments. Cancer Metab. 2016, 4, 6. [Google Scholar] [CrossRef] [PubMed]
  142. Kikuchi, K.; Tsukamoto, H. Stearoyl-CoA desaturase and tumorigenesis. Chem. Biol. Interact. 2020, 316, 108917. [Google Scholar] [CrossRef] [PubMed]
  143. Nagarajan, S.R.; Butler, L.M.; Hoy, A.J. The diversity and breadth of cancer cell fatty acid metabolism. Cancer Metab. 2021, 9, 2. [Google Scholar] [CrossRef] [PubMed]
  144. Kubota, C.S.; Espenshade, P.J. Targeting stearoyl-CoA desaturase in solid tumors. Cancer Res. 2022, 82, 1682–1688. [Google Scholar] [CrossRef]
  145. Raeisi, M.; Hassanbeigi, L.; Khalili, F.; Kharrati-Shishavan, H.; Yousefi, M.; Mehdizadeh, A. Stearoyl-CoA desaturase 1 as a therapeutic target for cancer: A focus on hepatocellular carcinoma. Mol. Biol. Rep. 2022, 49, 8871–8882. [Google Scholar] [CrossRef]
  146. Min, J.Y.; Kim, D. Stearoyl-CoA desaturase 1 as a therapeutic biomarker: Focusing on cancer stem cells. Int. J. Mol. Sci. 2023, 24, 8951. [Google Scholar] [CrossRef] [PubMed]
  147. Scaglia, N.; Igal, R.A. Stearoyl-CoA desaturase is involved in the control of proliferation, anchorage-independent growth, and survival in human transformed cells. J. Biol. Chem. 2005, 280, 25339–25349. [Google Scholar] [CrossRef]
  148. Scaglia, N.; Chisholm, J.W.; Igal, R.A. Inhibition of stearoylCoA desaturase-1 inactivates acetyl-CoA carboxylase and impairs proliferation in cancer cells: Role of AMPK. PLoS ONE 2009, 4, e6812. [Google Scholar] [CrossRef]
  149. Xuan, Y.; Wang, H.G.; Yung, M.M.H.; Chen, F.S.; Chan, W.S.; Chan, Y.S.; Tsui, S.K.W.; Ngan, H.Y.S.; Chan, K.K.L.; Chan, D.W. SCD1/FADS2 fatty acid desaturases equipoise lipid metabolic activity and redox-driven ferroptosis in ascites-derived ovarian cancer cells. Theranostics 2022, 12, 3534–3552. [Google Scholar] [CrossRef]
  150. Huang, J.; Fan, X.X.; He, J.X.; Pan, H.; Li, R.Z.; Huang, L.Y.; Jiang, Z.; Yao, X.-J.; Liu, L.; Leung, E.L.-H.; et al. SCD1 is associated with tumor promotion, late stage and poor survival in lung adenocarcinoma. Oncotarget 2016, 7, 39970–39979. [Google Scholar] [CrossRef]
  151. Pisanu, M.E.; Noto, A.; De Vitis, C.; Morrone, S.; Scognamiglio, G.; Botti, G.; Venuta, F.; Diso, D.; Jakopin, Z.; Padula, F.; et al. Blockade of stearoyl-CoA-desaturase 1 activity reverts resistance to cisplatin in lung cancer stem cells. Caner Lett. 2017, 406, 93–104. [Google Scholar] [CrossRef] [PubMed]
  152. Pisanu, M.E.; Maugeri-Sacca, M.; Fattore, L.; Bruschini, S.; De Vitis, C.; Tabbi, E.; Bellei, B.; Migliano, E.; Kovacs, D.; Camera, E.; et al. Inhibition of stearoyl-CoA desaturase 1 reverts BRAF and MEK inhibition-induced selection of cancer stem cells in BRAF-mutated melanoma. J. Exp. Clin. Cancer Res. 2018, 37, 318. [Google Scholar] [CrossRef] [PubMed]
  153. Hwang, S.H.; Yang, Y.; Jung, J.H.; Kim, Y. Oleic acid from cancer-associated fibroblast promotes cancer cell stemness by stearoyl-CoA desaturase under glucose-deficient condition. Cancer Cell Int. 2022, 22, 404. [Google Scholar] [CrossRef]
  154. Fritz, V.; Benfodda, Z.; Rodier, G.; Henriquet, C.; Iborra, F.; Avances, C.; Allory, Y.; de la Taille, A.; Culine, S.; Blancou, H.; et al. Abrogation of de novo lipogenesis by stearoyl-CoA desaturase 1 inhibition interferes with oncogenic signaling and blocks prostate cancer progression in mice. Mol. Cancer Ther. 2010, 9, 1740–1754. [Google Scholar] [CrossRef]
  155. Budhu, A.; Roessler, S.; Zhao, X.L.; Yu, Z.P.; Forgues, M.; Ji, J.F.; Karoly, E.; Qin, L.-X.; Ye, Q.-H.; Jia, H.-L.; et al. Integrated metabolite and gene expression profiles identify lipid biomarkers associated with progression of hepatocellular carcinoma and patient outcomes. Gastroenterology 2013, 144, 1066–1075. [Google Scholar] [CrossRef] [PubMed]
  156. Ma, M.K.F.; Lau, E.Y.T.; Leung, D.H.W.; Lo, J.; Ho, N.P.Y.; Cheng, L.K.W.; Ma, S.; Lin, C.H.; Copland, J.A.; Ding, J.; et al. Stearoyl-CoA desaturase regulates sorafenib resistance via modulation of ER stress-induced differentiation. J. Hepatol. 2017, 67, 979–990. [Google Scholar] [CrossRef]
  157. von Roemeling, C.A.; Marlow, L.A.; Pinkerton, A.B.; Crist, A.; Miller, J.; Tun, H.W.; Smallridge, R.C.; Copland, J.A. Aberrant lipid metabolism in anaplastic thyroid carcinoma reveals stearoyl CoA desaturase 1 as a novel therapeutic target. J. Clin. Endocrinol. Metab. 2015, 100, E697–E709. [Google Scholar] [CrossRef]
  158. Liu, G.H.; Feng, S.; Jia, L.; Wang, C.Y.; Fu, Y.; Luo, Y.Z. Lung fibroblasts promote metastatic colonization through upregulation of stearoyl-CoA desaturase 1 in tumor cells. Oncogene 2018, 37, 1519–1533. [Google Scholar] [CrossRef]
  159. Noto, A.; Raffa, S.; De Vitis, C.; Roscilli, G.; Malpicci, D.; Coluccia, P.; Di Napoli, A.; Ricci, A.; Giovagnoli, M.R.; Aurisicchio, L.; et al. Stearoyl-CoA desaturase-1 is a key factor for lung cancer-initiating cells. Cell Death Dis. 2013, 4, e947. [Google Scholar] [CrossRef]
  160. Noto, A.; De Vitis, C.; Pisanu, M.E.; Roscilli, G.; Ricci, G.; Catizone, A.; Sorrentino, G.; Chianese, G.; Taglialatela-Scafati, O.; Trisciuoglio, D.; et al. Stearoyl-CoA-desaturase 1 regulates lung cancer stemness via stabilization and nuclear localization of YAP/TAZ. Oncogene 2017, 36, 4573–4584. [Google Scholar] [CrossRef]
  161. Ran, H.; Zhu, Y.M.; Deng, R.Y.; Zhang, Q.; Liu, X.S.; Feng, M.; Zhong, J.; Lin, S.; Tong, X.; Su, Q. Stearoyl-CoA desaturase-1 promotes colorectal cancer metastasis in response to glucose by suppressing PTEN. J. Exp. Clin. Cancer Res. 2018, 37, 54. [Google Scholar] [CrossRef] [PubMed]
  162. Li, J.J.; Condello, S.; Thomes-Pepin, J.; Ma, X.X.; Xia, Y.; Hurley, T.D.; Matei, D.; Cheng, J.X. Lipid desaturation is a metabolic marker and therapeutic target of ovarian cancer stem cells. Cell Stem Cell 2017, 20, 303–318. [Google Scholar] [CrossRef] [PubMed]
  163. Bansal, S.; Berk, M.; Alkhouri, N.; Partrick, D.A.; Fung, J.J.; Feldstein, A. Stearoyl-CoA desaturase plays an important role in proliferation and chemoresistance in human hepatocellular carcinoma. J. Surg. Res. 2014, 186, 29–38. [Google Scholar] [CrossRef] [PubMed]
  164. Luo, H.; Wang, X.H.; Song, S.; Wang, Y.H.; Dan, Q.F.; Ge, H. Targeting stearoyl-CoA desaturase enhances radiation induced ferroptosis and immunogenic cell death in esophageal squamous cell carcinoma. Oncoimmunology 2022, 11, 2101769. [Google Scholar] [CrossRef]
  165. Zhang, R.H.; Liu, L.G.; Wang, F.Q.; Zhao, W.Z.; Liu, K.; Yu, H.; Zhao, S.; Xu, B.; Zhang, X.; Chai, J.; et al. AKAP8L enhances the stemness and chemoresistance of gastric cancer cells by stabilizing SCD1 mRNA. Cell Death Dis. 2022, 13, 1041. [Google Scholar] [CrossRef] [PubMed]
  166. Morais, C.M.; Cardoso, A.M.; Araujo, A.R.D.; Reis, A.; Domingues, P.; Domingues, M.R.M.; de Lima, M.C.P.; Jurado, A.S. Stearoyl CoA desaturase-1 silencing in glioblastoma cells: Phospholipid remodeling and cytotoxicity enhanced upon autophagy inhibition. Int. J. Mol. Sci. 2022, 23, 13014. [Google Scholar] [CrossRef]
  167. Parik, S.; Fernandez-Garcia, J.; Lodi, F.; De Vlaminck, K.; Derweduwe, M.; De Vleeschouwer, S.; Sciot, R.; Geens, W.; Weng, L.; Bosisio, F.M.; et al. GBM tumors are heterogeneous in their fatty acid metabolism and modulating fatty acid metabolism sensitizes cancer cells derived from recurring GBM tumors to temozolomide. Front. Oncol. 2022, 12, 988872. [Google Scholar] [CrossRef]
  168. Lien, E.C.; Westermark, A.M.; Zhang, Y.; Yuan, C.; Li, Z.Q.; Lau, A.N.; Sapp, K.M.; Wolpin, B.M.; Vander Heiden, M.G. (Low glycaemic diets alter lipid metabolism to influence tumour growth. Nature 2021, 599, 302–307. [Google Scholar] [CrossRef]
  169. Sun, M.; Chen, X.X.; Yang, Z.B. Single cell mass spectrometry studies reveal metabolomic features and potential mechanisms of drug-resistant cancer cell lines. Anal. Chim. Acta 2022, 1206, 339761. [Google Scholar] [CrossRef]
  170. Li, W.; Bai, H.; Liu, S.; Cao, D.; Wu, H.; Shen, K.; Tai, Y.; Yang, J. Targeting stearoyl-CoA desaturase 1 to repress endometrial cancer progression. Oncotarget 2018, 9, 12064–12078. [Google Scholar] [CrossRef]
  171. Piao, C.Y.; Cui, X.L.; Zhan, B.; Li, J.; Li, Z.L.; Li, Z.H.; Liu, X.; Bi, J.; Zhang, Z.; Kong, C.Z. Inhibition of stearoyl CoA desaturase-1 activity suppresses tumour progression and improves prognosis in human bladder cancer. J. Cell. Mol. Med. 2019, 23, 2064–2076. [Google Scholar] [CrossRef] [PubMed]
  172. Wang, J.F.; Xu, Y.Z.; Zhe, L.S.; Zou, Y.; Kong, W.; Dong, B.J.; Huang, J.; Chen, Y.; Xue, W.; Zhang, J. High expression of stearoyl-CoA desaturase 1 predicts poor prognosis in patients with clear cell renal cell carcinoma. PLoS ONE 2016, 11, e0166231. [Google Scholar] [CrossRef] [PubMed]
  173. Ducheix, S.; Peres, C.; Hardfeldt, J.; Frau, C.; Mocciaro, G.; Piccinin, E.; Lobaccaro, J.-M.; De Santis, S.; Chieppa, M.; Bertrand-Michel, J.; et al. Deletion of stearoyl-CoA desaturase-1 from the intestinal epithelium promotes inflammation and tumorigenesis, reversed by dietary oleate. Gastroenterology 2018, 155, 1524–1532. [Google Scholar] [CrossRef] [PubMed]
  174. Gunes, B.A.; Hekmatshoar, Y.; Ozkan, T.; Bozkurt, S.; Aydos, O.S.E.; Buyukasik, Y.; Aladag, E.; Sunguroglu, A. Downregulation of stearoyl-CoA desaturase 1 (SCD-1) promotes resistance to imatinib in chronic myeloid leukemia. Mediterr. J. Hematol. Infect. Dis. 2023, 15, e2023008. [Google Scholar] [CrossRef]
  175. Van den Branden, C.; Roels, F. Thioridazine: A selective inhibitor of peroxisomal beta-oxidation in vivo. FEBS Lett. 1985, 187, 331–333. [Google Scholar] [CrossRef]
  176. Shi, R.L.; Zhang, Y.; Shi, Y.; Shi, S.Y.; Jiang, L.L. Inhibition of peroxisomal beta-oxidation by thioridazine increases the amount of VLCFAs and A beta generation in the rat brain. Neurosci. Lett. 2012, 528, 6–10. [Google Scholar] [CrossRef]
  177. Beach, S.R.; Celano, C.M.; Noseworthy, P.A.; Januzzi, J.L.; Huffman, J.C. QTc prolongation, torsades de pointes, and psychotropic medications. Psychosomatics 2013, 54, 1–13. [Google Scholar] [CrossRef]
  178. Huang, X.; Gan, G.M.; Wang, X.X.; Xu, T.; Xie, W. The HGF-MET axis coordinates liver cancer metabolism and autophagy for chemotherapeutic resistance. Autophagy 2019, 15, 1258–1279. [Google Scholar] [CrossRef]
  179. Yu, D.L.; Li, Y.T.; Sun, K.D.Y.; Gu, J.J.; Chen, Z.; Owonikoko, T.K.; Ramalingam, S.S.; Sun, S.Y. The novel MET inhibitor, HQP8361, possesses single agent activity and enhances therapeutic efficacy of AZD9291 (osimertinib) against AZD9291-resistant NSCLC cells with activated MET. Am. J. Cancer Res. 2020, 10, 3316–3327. [Google Scholar]
  180. Luttich, L.; Besso, M.J.; Heiden, S.; Koi, L.; Baumann, M.; Krause, M.; Dubrovska, A.; Linge, A.; Kurth, I.; Peitzsch, C. Tyrosine kinase c-MET as therapeutic target for radiosensitization of head and neck squamous cell carcinomas. Cancers 2021, 13, 1865. [Google Scholar] [CrossRef]
  181. Oh, H.N.; Kwak, A.W.; Lee, M.H.; Kim, E.; Yoon, G.; Cho, S.S.; Liu, K.; Chae, J.-I.; Shim, J.H. Targeted inhibition of c-MET by podophyllotoxin promotes caspase-dependent apoptosis and suppresses cell growth in gefitinib-resistant non-small cell lung cancer cells. Phytomedicine 2021, 80, 153355. [Google Scholar] [CrossRef] [PubMed]
  182. Terlecka, P.; Krawczyk, P.; Grenda, A.; Milanowski, J. MET gene dysregulation as a promising therapeutic target in lung cancer—A review. J. Pers. Med. 2021, 11, 1370. [Google Scholar] [CrossRef] [PubMed]
  183. Yu, Y.; Peng, X.D.; Qian, X.J.; Zhang, K.M.; Huang, X.; Chen, Y.H.; Li, Y.-T.; Feng, G.-K.; Zhang, H.-L.; Xu, X.-L.; et al. Fis1 phosphorylation by Met promotes mitochondrial fission and hepatocellular carcinoma metastasis. Signal Transduct. Target. Ther. 2021, 6, 401. [Google Scholar] [CrossRef]
  184. Chen, Z.; Vallega, K.A.; Chen, H.Y.; Zhou, J.; Ramalingam, S.S.; Sun, S.Y. The natural product berberine synergizes with osimertinib preferentially against MET-amplified osimertinib-resistant lung cancer via direct MET inhibition. Pharmacol. Res. 2022, 175, 105998. [Google Scholar] [CrossRef]
  185. Takumi, Y.; Arai, S.; Suzuki, C.; Fukuda, K.; Nishiyama, A.; Takeuchi, S.; Sato, H.; Matsumoto, K.; Sugio, K.; Yano, S. MET kinase inhibitor reverses resistance to entrectinib induced by hepatocyte growth factor in tumors with NTRK1 or ROS1 rearrangements. Cancer Med. 2023, 12, 5809–5820. [Google Scholar] [CrossRef]
  186. Xu, X.M.; Yao, L. Recent patents on the development of c-MET kinase inhibitors. Recent Pat. Anti Cancer Drug Discov. 2020, 15, 228–238. [Google Scholar] [CrossRef] [PubMed]
  187. Dai, J.T.; Zhang, J.Q.; Fu, D.X.; Liu, M.; Zhang, H.; Tang, S.; Wang, L.; Xu, S.; Zhu, W.; Tang, Q.; et al. Design, synthesis and biological evaluation of 4-(4-aminophenoxy) picolinamide derivatives as potential antitumor agents. Eur. J. Med. Chem. 2023, 257, 115499. [Google Scholar] [CrossRef]
  188. Michaelides, I.N.; Collie, G.W.; Borjesson, U.; Vasalou, C.; Alkhatib, O.; Barlind, L.; Cheung, T.; Dale, I.L.; Embrey, K.J.; Hennessy, E.J.; et al. Discovery and optimization of the first ATP competitive type-III c-MET inhibitor. J. Med. Chem. 2023, 13, 8782–8807. [Google Scholar] [CrossRef]
  189. Zeng, X.C.; Hu, Y.W.; Xu, Z.; Wu, X.Y.; Xiong, Y.; Liu, S.P. Development of new c-MET inhibitors and application of corresponding multitarget tyrosine kinase inhibitors in tumor therapy. Soft Comput. 2023; early access. [Google Scholar] [CrossRef]
  190. Ovens, M.J.; Manoharan, C.; Wilson, M.C.; Murray, C.M.; Halestrap, A.P. The inhibition of monocarboxylate transporter 2 (MCT2) by AR-C155858 is modulated by the associated ancillary protein. Biochem. J. 2010, 431, 217–225. [Google Scholar] [CrossRef]
  191. Curtis, N.J.; Mooney, L.; Hopcroft, L.; Michopoulos, F.; Whalley, N.; Zhong, H.H.; Murray, C.; Logie, A.; Revill, M.; Byth, K.F.; et al. Pre-clinical pharmacology of AZD3965, a selective inhibitor of MCT1: DLBCL, NHL and Burkitt’s lymphoma anti-tumor activity. Oncotarget 2017, 8, 69219–69236. [Google Scholar] [CrossRef]
  192. Singh, M.; Afonso, J.; Sharma, D.; Gupta, R.; Kumar, V.; Rani, R.; Baltazar, F.; Kumar, V. Targeting monocarboxylate transporters (MCTs) in cancer: How close are we to the clinics? Semin. Cancer Biol. 2023, 90, 1–14. [Google Scholar] [CrossRef] [PubMed]
  193. Powell, D.A. An overview of patented small molecule stearoyl coenzyme-A desaturase inhibitors (2009–2013). Expert Opin. Ther. Pat. 2014, 24, 155–175. [Google Scholar] [CrossRef] [PubMed]
  194. Imamura, K.; Tomita, N.; Kawakita, Y.; Ito, Y.; Ono, K.; Nii, N.; Miyazaki, T.; Yonemori, K.; Tawada, M.; Sumi, H.; et al. Discovery of novel and potent stearoyl Coenzyme A desaturase 1 (SCD1) inhibitors as anticancer agents. Bioorganic Med. Chem. 2017, 25, 3768–3779. [Google Scholar] [CrossRef]
  195. Meimaridou, E.; Kowalczyk, J.; Guasti, L.; Hughes, C.R.; Wagner, F.; Frommolt, P.; Nuernberg, P.; Mann, N.P.; Banerjee, R.; Saka, H.N.; et al. Mutations in NNT encoding nicotinamide nucleotide transhydrogenase cause familial glucocorticoid deficiency. Nat. Genet. 2012, 44, 740–742. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Key processes in the “lipid metabolism resistance system” (LMRS). In orange text are enzyme activities managing the specialized catabolic flow of fatty acids found in many advanced carcinomas (text). One of the central assumptions of this picture is that this system produces elevated lipid catabolism, engendering increased levels of energy and reducing potential under the conditions of in vivo carcinomas, including inadequate oxygen supplies. These increases, in turn, are proposed to allow carcinoma cells to better survive and recover from therapeutic assault, thereby rendering them broadly treatment-resistant. (IMM and OMM = inner and outer mitochondrial membranes.)
Figure 1. Key processes in the “lipid metabolism resistance system” (LMRS). In orange text are enzyme activities managing the specialized catabolic flow of fatty acids found in many advanced carcinomas (text). One of the central assumptions of this picture is that this system produces elevated lipid catabolism, engendering increased levels of energy and reducing potential under the conditions of in vivo carcinomas, including inadequate oxygen supplies. These increases, in turn, are proposed to allow carcinoma cells to better survive and recover from therapeutic assault, thereby rendering them broadly treatment-resistant. (IMM and OMM = inner and outer mitochondrial membranes.)
Ijms 24 14365 g001
Table 1. In light of the large body of evidence for LMRS metabolism involvement in advanced carcinoma treatment resistance, assessing opportunities for clinical deployment of these insights is important. Several studies have been conducted that support such assessments and possible planning for future clinical trials. This table briefly summarizes the most useful of these of which we are aware.
Table 1. In light of the large body of evidence for LMRS metabolism involvement in advanced carcinoma treatment resistance, assessing opportunities for clinical deployment of these insights is important. Several studies have been conducted that support such assessments and possible planning for future clinical trials. This table briefly summarizes the most useful of these of which we are aware.
ReferenceCarcinomaFundamental Observation
IN VITRO (cell culture)
[151]lung carcinomaMF-438 inhibition of SCD1 desaturase reduces cisplatin resistance in lung carcinoma 3D spheroids
[152]melanomaMF-438 inhibition of SCD1 desaturase reduces resistance to BRAFi plus MEKi inhibition of melanoma 3D spheroid formation
[167]Glioblastoma(GBMMerck inhibitors of SCD1 (Cpd3j) or FADS2 (SC26196) desaturases sensitize GBM cells to temozolomide-induced cell death in otherwise resistant cell lines
IN VIVO (tumor models)
[30]melanomaEtomoxir fatty acid oxidation inhibitor interferes with nongenetic xenograft resistance to BRAF inhibitors (in the presence of DCA perturbation of glycolytic regulation).
[119]melanomaThioridazine fatty acid oxidation inhibitors robustly block nongenetic xenograft resistance to combinations of BRAF and MEK inhibitors.
[51]pancreatic adenocarcinoma(PDAC)Orlistat inhibition of fatty acid synthesis robustly blocks gemcitabine resistance in orthotopic xenografts.
[121]pancreatic adenocarcinoma(PDAC)Thioridazine substantially interferes with xenograft resistance to tumor-specific TCA cycle inhibitor, CPI-613. Crizotinib METi mimics this in vivo thioridazine effect, apparently through LMRS interference.
[156]hepatocellular carcinoma (HCC)Novel inhibitor (SSI-4) suppression of SCD1 activity robustly overcomes HCC xenograft resistance to multi-RTK inhibitor, sorafinib.
[164]esophageal squamous cell carcinoma(ESCC)MF-438 SCD1 inhibitor moderately enhances ESCC xenograft radiation sensitivity.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bingham, P.M.; Zachar, Z. Toward a Unifying Hypothesis for Redesigned Lipid Catabolism as a Clinical Target in Advanced, Treatment-Resistant Carcinomas. Int. J. Mol. Sci. 2023, 24, 14365. https://doi.org/10.3390/ijms241814365

AMA Style

Bingham PM, Zachar Z. Toward a Unifying Hypothesis for Redesigned Lipid Catabolism as a Clinical Target in Advanced, Treatment-Resistant Carcinomas. International Journal of Molecular Sciences. 2023; 24(18):14365. https://doi.org/10.3390/ijms241814365

Chicago/Turabian Style

Bingham, Paul M., and Zuzana Zachar. 2023. "Toward a Unifying Hypothesis for Redesigned Lipid Catabolism as a Clinical Target in Advanced, Treatment-Resistant Carcinomas" International Journal of Molecular Sciences 24, no. 18: 14365. https://doi.org/10.3390/ijms241814365

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop