Abstract
Reprogramming of cellular metabolism is a hallmark of cancer. Mitochondrial ATP synthase (MAS) produces most of the ATP that drives the cell. High expression of the MAS-composing proteins is found during cancer and is linked to a poor prognosis in glioblastoma, ovarian cancer, prostate cancer, breast cancer, and clear cell renal cell carcinoma. Cell surface-expressed ATP synthase, translocated from mitochondrion to cell membrane, involves the angiogenesis, tumorigenesis, and metastasis of cancer. ATP synthase has therefore been considered a therapeutic target. We review recent various ATP synthase inhibitors that suppress tumor growth and are being tested for the clinic.
Keywords: mitochondrial ATP synthase, cancer metabolism, mitochondrial metabolism, ATP synthase inhibitor, chemotherapy
Graphical abstract

The review highlights the ongoing development of ATP synthase inhibitors that have shown effectiveness in the treatment of cancer. High expression of the ATP synthase composing proteins is found during cancer and links to a poor prognosis in various tumors. Many ATP synthase inhibitors suppress tumor growth and are being tested for the clinic.
Introduction
A century ago, Warburg reported that the metabolism in cancer has changed.1 Reprogramming of metabolism is one of the hallmarks of cancer.2 Drugs that target this reprogramming are emerging as novel drug families, and some are being evaluated in clinical trials (Figure 1).3 The mitochondrion plays a key role in the metabolism reprogrammed in cancer.3, 4, 5 For example, cancer cells without mitochondria can only form tumors if they reconstitute oxidative phosphorylation (OXPHOS) from host stroma.6 The mitochondrion is also linked to the resistance of tumor stem cells to treatment and in pancreatic cancer to KRAS-driven signaling inhibition.7, 8, 9
Figure 1.
Different therapeutic strategies in cancer treatment
ATP synthase as a novel target among different therapeutic strategies of cancer.
Mitochondrial ATP synthase (MAS), an enzyme that produces ATP, also known as complex V, is the final and pivotal enzyme of OXPHOS in the mitochondrion.10 In 1994, ectopically expressed ATP synthase (ectopic ATP synthase [EAS]) on the cell surface of vascular endothelial cells and tumor cells was found.11 The existence of EAS is reportedly related to tumor angiogenesis, metastasis, and drug resistance.12, 13, 14, 15
We review the ongoing development of ATP synthase inhibitors that have shown effectiveness in the treatment of cancer.16
The structure of MAS
MAS is the primary generator of ATP and is widely found in the cell membranes of bacteria, the thylakoid membranes of chloroplasts in algae, and the inner membranes of the mitochondria of eukaryotic cells.17 MAS transduces the electrochemical gradient of the proton motive force across these membranes into ATP, which is generated during respiration (Figure 2).
Figure 2.
Schematic diagram of the how MAS works in mitochondrion
The respiratory chain builds up proton motive force, and ATP is produced by MAS. I, complex I; II, complex II; III, complex III; IV, complex IV; Q, coenzyme Q.
Its complex structure has been elucidated by electron microscopy.18, 19, 20 The mammalian MAS consists of 28 subunits from 17 polypeptides, assembled into (1) a membrane-bound rotor and (2) a catalytic, ATP-generating center in the cytoplasm, linked by a central stalk and a peripheral stalk (PS).18 Protons traverse the membrane inside the rotor, which combined with the PS is called the Fo region; the F1 region consists of the ATP-generating catalytic crown and its asymmetrical central stalk (Figure 3).
Figure 3.
Current view of the structure of ATP synthase
Different subunits are shown by different colors, and their names are on the right linked by black lines.
The rotor consists of a ring of eight c-subunits, clung by an ATP6 subunit. The PS part includes the subunits of the oligomycin sensitivity conferring protein (OSCP), b, d, and F6. Subunits e, f, g, ATP8, and 6.8 proteolipid are the accessory subunits of the PS.
The catalytic crown harbors three β subunits, the catalytic sites, and three noncatalytic α subunits. These β subunits can undergo three different conformations: (1) an open state with no nucleotide bound (E), (2) the loose state with ADP and Pi bound (D), and (3) the tight state with synthesized or hydrolyzed bound ATP (T). With the rotation of the c-ring, the conformation of catalytic sites alternates from E to T and D states. The asymmetrical stalk, composed of subunits γ, δ, and ε, connects the Fo inner membrane domain and the F1 catalytic domain.
The monomeric MASs dimerize via interaction between ATP6 subunits and between 6.8 proteolipids, and then the dimers are linked together by a DAPIT subunit along the edge of the cristae.20,21
ATP synthase is required for malignant tumor growth
MAS in tumor
MAS relates to cancer through mutation of the genes coding for its subunits and also through abnormal expression of genes coding for these subunits.14,22, 23, 24, 25 In glioblastoma, the mRNA levels of the MAS F1 subunits α and β (ATP5A1, and ATP5B) are significantly higher in tumor cells and endothelial cell of microvascular proliferation, attributed to the downregulation of microRNAs (miRNAs) that target ATP5A1 and ATP5B.26
ATP5B
In ovarian cancer, downregulated miR-450a that targets ATP5B expression increases metastasis, indicating that ATP5B might support the invasiveness of ovarian cancer.27 In prostate cancer, breast cancer, and thyroid cancer,28,29 overexpressed ATP5B negatively correlates with metastasis-free and overall survival. HER2+ breast tumors that had acquired resistance to HER2-targeted therapies showed increased expression of ATP5B and ATP synthase PS subunit F6 (ATP5J).26
ATP5A1
In clear cell renal cell carcinoma and in breast cancer progression and metastasis, ATP5A1’s overexpression is associated with progression, a potential biomarker for diagnosis and prognosis and the treatment response.30,31 In prostate cancer, ATP5A1 levels positively correlate with the early onset of tumor.32 In colorectal cancer, high expression of ATP5A1 is associated with SNPs, TP53 mutation, and chromosomal instability to facilitate tumor development.33 In hepatocellular cancer, ATP5A1 also fell in the ingredient-target network of dihydroartemisinin and was responsible for the anti-hepatocellular carcinoma effect.34 In lung cancer, periplocin inhibits growth of lung cancer by downregulating ATP5A1 and other key proteins.35
ATP level alteration also reflects the development of carcinogenesis,36 marking the tumor region and its microenvironment.37,38 Moreover, MAS is able to modulate the astrocyte inflammatory response, which is related to the initiation of glioma.39,40
Additionally, mutations of MAS-coding genes are accumulated with the carcinogenesis process, suggesting the participation of MAS in the biological process of cancer cells. Two mutations in MT-ATP6, which encodes subunit ATP6 of MAS, affect the calcium homeostasis and activation of the permeability transition pore (PTP) in yeast,23 the latter of which is prominent for cancer cells to escape from apoptosis.
EAS in tumor
EAS is located not only on the mitochondrial inner membrane, but also on the plasma membrane of tumor cells and human vascular endothelial cells.11,41 EAS is the receptor of angiostatin in endothelial cells, and it might be involved in maintaining the normal intracellular pH of cancer cells in an acidic extracellular microenvironment.12 It is translocated from mitochondria to cell surface, and mitochondrial transit peptide is essential for ATP synthase localization on the cell surface and mitochondria.13 In hepatocellular carcinoma (HCC), blocking EAS could inhibit extracellular ATP synthesis and have antiangiogenic and antitumorigenic activities.14 In lung cancer, the level of EAS is correlated with gefitinib sensitivity and cell survival.15 In leukemia, ATP5B of EAS on the human leukemia cell surface interacts with the platelet GPIIb epitope, causing platelet-cancer aggregation, with the latter serving as a target for a mimetic cytotoxic peptide to be specifically delivered to ectoATP5B-positive cells, contributing to targeted cancer cell killing.42
Inhibiting the activity of ATP synthase can effectively impair tumor metabolism reprogramming and tumor growth.43 ATP synthase is essential for tumor progression and has potential as a target in cancer treatment.
Cancer therapy based on targeting ATP synthase
Given the role of ATP synthase in fueling cancer, ATP synthase inhibitors have been explored as a new cancer therapy.44,45 Categories that have been developed and investigated are polyketide inhibitors, polyphenolic phytochemicals, estrogens, polyenic α-pyrone derivatives, synthetic compounds, and protein peptides,46,47 some of which were earlier used as antibiotics to treat infectious diseases or anti-obesity drugs and later showed antiangiogenetic and anticancer activity.48, 49, 50
AS inhibitor development is at the start of its debut in cancer treatment. None has become commercially available for clinical cancer treatment. Table 1 summarizes current inhibitors under investigation, sources, inhibitory sites, and related studies in tumor treatment (see Figure 4 and Table 1). Figure 5 displays the structure of AS inhibitors.
Table 1.
ATP synthase inhibitors as anticancer drugs
| Name | Source | Mode of action | Diseases | Research stage | Refs. |
|---|---|---|---|---|---|
| Polyketides | |||||
| Oligomycin | Streptomyces | binding to the Fo region | liver cancer, non-small cell lung cancer, colon cancer, triple-negative breast cancer, ovarian cancer | preclinical | 51, 52, 53 |
| Apoptolidin | Nocardiopsis spp. | binding to the Fo region | lung cancer | preclinical | 54,55 |
| Polyphenolics | |||||
| Resveratrol | grapes and berries | binding a hydrophobic pocket between the C-terminal region of the γ subunit and the hydrophobic inside of the β subunit | pheochromocytoma colon cancer | phase I in colon cancer (completed): ClinicalTrials.gov: NCT00256334, NCT00920803, and NCT00433576; phase I in unspecified adult solid tumor (completed): ClinicalTrials.gov: NCT00098969; phase II in multiple myeloma (terminated): ClincialTrials.gov: NCT00920556; phase I in polycystic ovary syndrome (terminated): ClinicalTrials.gov: NCT01489319 | 56, 57, 58 |
| EGCG | green tea | inhibiting the ATP hydrolysis activity of ATP synthase | hepatocellular carcinoma, malignant pleural mesothelioma, colon cancer | early phase I in colon cancer (recruiting): ClinicalTrials.gov: NCT02891538; phase I & II in prostate cancer: ClinicalTrials.gov: NCT00459407 (completed) and NCT04300855 (recruiting); phase I & II in breast cancer: ClinicalTrials.gov: NCT00516243 (completed), NCT02580279 (unknown), and NCT04597359 (not yet recruiting); phase II in lung cancer (enrolling by invitation): ClinicalTrials.gov: NCT02577393; phase II in nonmetastatic bladder cancer (completed): ClinicalTrials.gov: NCT00666562. | 59, 60, 61 |
| Curcumin | the roots and stalks of Zingiberaceae | inhibiting the activity of ATP synthase | liver cancer, breast cancer, cervical carcinoma, prostatic cancer, lung cancer, leukemia, melanoma, colon cancer | phase II in breast cancer (completed): ClinicalTrials.gov: NCT01042938; phase III in prostate cancer (recruiting): ClinicalTrials.gov: NCT03769766; phase II in colorectal cancer (completed): ClinicalTrials.gov: NCT02439385, and so on | 43,62, 63, 64, 65, 66, 67, 68 |
| 17β-estradiol | Homo sapiens | binding to the OSCP subunit of Fo region | liver tissue, rat brain | phase II in breast cancer: ClinicalTrials.gov: NCT01083641(terminated, has result), NCT00324259 (completed), and NCT01385280 (completed); phase II in prostate cancer (terminated, has result): ClinicalTrials.gov: NCT00459810; phase I in solid cancers (completed): ClinicalTrials.gov: NCT01209143; phase I in hematologic malignancies (recruiting): ClinicalTrials.gov: NCT03557619, and so on | 69, 70, 71, 72, 73, 74 |
| Genistein | Soybean | targeting polyphenol binding pocket of ATP synthase and blocking the rotation of the γ subunit | breast cancer | phase II in breast cancer (completed): ClinicalTrials.gov: NCT00244933 and NCT00290758; phase II & III in prostate cancer (completed): ClinicalTrials.gov: NCT00584532 and NCT01325311; phase I & II in non-small cell lung cancer (completed): ClinicalTrials.gov: NCT01628471 and NCT02567799; phase I & II in colon cancer (completed): ClinicalTrials.gov: NCT01985763; phase I in breast cancer & endometrial cancer (completed): ClinicalTrials.gov: NCT00099008; phase II in bladder cancer (recruiting): ClinicalTrials.gov: NCT01489813 and NCT00118040; phase II in pancreatic cancer (completed): ClinicalTrials.gov: NCT00376948; phase I & II in pediatric relapsed or refractory malignancies (completed): ClinicalTrials.gov: NCT02499861; early phase I in kidney cancer & melanoma (completed): ClinicalTrials.gov: NCT00276835; phase I in cancer of head and neck (completed): ClinicalTrials.gov: NCT02075112 | 75,76 |
| Polyenic a-pyrones | |||||
| Citreoviridin | Penicillium, Aspergillus | binding to the β subunit of the F1 region | lung adenocarcinoma, breast cancer | preclinical | 41,77, 78, 79 |
| Aurovertin B | Calcarisporium arbuscular | binding to the β subunit of the F1 region | breast cancer, colon cancer | preclinical | 79, 80, 81 |
| Asteltoxin | Aspergillus stellatus Curzi and Emericella variecolor | binding to the β subunit of the F1 region | lung cancer, breast cancer | preclinical | 82,83 |
| Synthetics | |||||
| Bedaquiline | synthetic | binding to the c subunit rotor and subunit ε of the ATP synthase | tuberculosis, lung cancer, glioblastoma, breast cancer | preclinical | 84, 85, 86, 87, 88, 89, 90 |
| Gboxin | synthetic | its positive charge influences the proton gradient of the inner mitochondrial membrane | glioblastoma | preclinical | 5 |
| Pd(II)COS@GbA | synthetic | inducing mitochondrial fragmentation and disrupting ATP synthase action | prostate cancer | preclinical | 91 |
| Edelfosine | synthetic | dissipation of the mitochondrial membrane potential | cervix epithelioid carcinoma, hematopoietic cancer | preclinical | 92, 93, 94, 95 |
| Miltefosine | synthetic | dissipation of the mitochondrial membrane potential | breast cancer | approved in the topical treatment of metastatic skin lesions in breast cancer | 96 |
| Perifosine | synthetic | dissipation of the mitochondrial membrane potential | advanced solid and leukemic tumors, brain tumors | phase I in advanced solid and leukemic tumors | 97,98 |
| Proteins | |||||
| IF1 | Homo sapiens | interacting with the F1 domain at the catalytic surface between the α and β subunit of ATP synthase, blocking the rotation of the complex | bladder carcinoma, glioma, metastatic colon cancer, triple-negative breast cancer | preclinical | 10,99, 100, 101 |
| Angiostatin | Homo sapiens | binding to α/β subunits of ATP synthase | colon carcinoma | phase I in unspecified adult solid tumor (completed): ClinicalTrials.gov: NCT00086723; phase II in non-small cell lung cancer (completed): ClinicalTrials.gov: NCT00049790 | 102 |
| Hai178 | humanized antibody | targeting ATP synthase | breast cancer, prostate cancer, lung cancer | preclinical | 103 |
EGCG, epigallocatechin gallate; OSCP, oligomycin sensitivity conferring protein; Pd(II)COS@GbA, Pd(II) complex of Gboxin analog-chitooligosaccharides conjugate; IF1, ATP synthase inhibitory factor 1.
Figure 4.
Inhibitory sites of ATP synthase
Different ATP synthase inhibitors targets different subunits, and the inhibitory sites are indicated by black lines.
Figure 5.
Structure of MAS inhibitors
EGCG, epigallocatechin gallate; Pd(II)COS@GbA, Pd(II) complex of Gboxin analog-chitooligosaccharides conjugate; IF1, ATP synthase inhibitory factor 1. The structures of inhibitors were obtained from the ChemSpider database.
Polyketide inhibitors
Polyketides are bioactive natural products isolated from diverse microorganisms, polymers of two ketides synthesized by polyketide synthase.104 Macrolides such as oligomycin, apoptolidin, and cytovaricin are polyketides and are MAS inhibitors with an anti-tumor activity.
Oligomycin
This compound, produced by Streptomyces strains, is a specific ATP synthase inhibitor that binds to the c subunit of Fo, as well at high concentrations to the F1 region,51,105 inhibiting ATP synthesis. The subunit oligomycin sensitivity-conferring protein (OSCP) is required for MAS sensitivity to oligomycin.106 Oligomycin can suppress cell viability in multiple cancers. It reduces the viability of hepatocellular carcinoma and non-small cell lung cancer cells with upregulated OXPHOS-like high-level SALL4 expression in vitro and in vivo.52 In colon cancer and triple-negative breast carcinoma it decreases cell proliferation, migration capacity, and invasiveness, revealing the potential for therapy.53,107 Oligomycin also significantly suppressed the survival and metastasis of microsatellite-stable colorectal cancer cells with increased mitochondrial DNA copy number.108
Moreover, oligomycin exhibits antitumor efficiency, particularly in tumors showing resistance to corresponding target therapies. It reverses the acquired resistance of HER2+ breast tumors to HER2-targeted therapies and enhances the antitumor efficacy of c-MET inhibition in glioblastoma cells; it also prevents recurrence in ovarian cancer.25,109,110 Inhibitors of OXPHOS, such as oligomycin, might be used for combined treatments.
Oligomycin, ossamycin, apoptolidin, and cytovaricin belong to the class of polyketide inhibitors of MAS with antifungal activities.46 The former three macrolide inhibitors belong to the top 0.1% cytotoxic agents in 37,000 small molecules tested against 60 human cancer cell lines.111
Apoptolidin
This compound, including isoforms A–D, are microbial secondary metabolites that highly selectivity induce apoptosis in some cancer cell lines.54,55,112 MAS is one of the biological targets for apoptolidins to exert antiproliferative activity.113 In a study by Ishmael and colleagues,114 apoptolidins A and C acted as AMPK activators and triggered autophagy in sensitive cell types without significant inhibition of mTORC1, showing that all macrolides do not function similarly.
Cytovaricin B
This compound was screened as an inhibitor of the JAK-STAT signaling pathway 20 years ago, and extended effects of this compound on tumor cells are being explored.115
Polyphenolic phytochemicals and estrogen
Polyphenolic phytochemicals are natural compounds from plants that contain phenol groups and show antitumor properties.116 Many inhibit ATP synthase. The number of phenol groups and the position of the hydroxyl groups affect the strength of the inhibition.117
Resveratrol and piceatannol
These two polyphenolic compounds are naturally found in diverse plants, mainly such as grapes, berries, and white tea. They are phytoestrogens that possess antioxidant, anti-inflammatory, cardioprotective, and anti-cancer properties.56,57 Resveratrol and piceatannol provide a wide range of preventive and therapeutic options against different types of cancer.57 They bind to a hydrophobic pocket between the hydrophobic C terminal region of the γ subunit and the hydrophobic inside of the β subunit.46 In this way, resveratrol promotes human pheochromocytoma cell death.58
EGCG
One of the catechins, epigallocatechin gallate (EGCG), is abundant in green tea and inhibits ATP hydrolysis.46 EGCG facilitates apoptosis as well as cell cycle arrest, growth inhibition in hepatocellular carcinoma, malignant pleural mesothelioma cells, and bladder urothelial cells.59, 60, 61 EGCG can prevent various chronic diseases, including neurodegenerative disorders, through several molecular signaling pathways including MAS inhibition.118
Curcumin
This MAS inhibitor is a natural polyphenol extracted from the roots and stalks of Zingiberaceae Curcuma longa, an herbal medicine with antitumor activity.62,119 It inhibits many cancers, including liver cancer, breast cancer, cervical carcinoma, prostatic cancer, and lung cancer. It affects several signaling pathways, with nuclear factor κB (NF-κB) signaling being prominently inhibited.63, 64, 65, 66, 67, 68 Recently MAS inhibition independent of NF-κB signaling was reported, which lowered ATP levels concomitantly with oxygen consumption, both in vivo and in vitro.
The energetic impairment by curcumin of L1210 murine lymphocytic leukemia, 4T1 murine breast, B16 murine melanoma, and CT26 murine colon tumor cell lines translated, also both in vitro and in vivo, into decreased tumor growth.44 According to ClinicalTrials.gov, 69 clinical studies have been done on curcumin, with 28 completed clinical trials, including 14 phase II trials. Curcumin on its own or in combination has been applied in studies of primary or metastatic breast cancer, colorectal cancer, pancreatic cancer, and endometrial carcinoma.
17β-estradiol (E2)
This compound plays a well-known role in breast cancer, binding to the estrogen receptor. In liver cells it also binds to the OSCP protein of MAS, promoting uncoupling.69, 70, 71, 72,120
Genistein
This isoflavone phytoestrogen from soybeans noncompetitively inhibits MAS. It targets the polyphenol binding pocket and blocks the rotation of the γ subunit.75 Both genistein and estradiol induce cell proliferation and apoptosis inhibition, cell cycle arrest and mitochondrial function, and dynamics damage of breast cancer cell lines.76
At present, 353 clinical studies of estradiol have been published. 61 clinical trials have been completed with results, and three clinical trials (phase I and phase II) have been completed. The drug was applied in the treatment of estrogen receptor-positive breast cancer, recurrent breast cancer, or solid cancers (ClinicalTrials.gov: NCT00324259, NCT01385280, and NCT01209143).
Polyenic α-pyrones
A polyenic α-pyrone is a six-membered cyclic unsaturated ester present in many natural products. Citreoviridin, aurovertin B and asteltoxin are all polyenic α-pyrone, and they target the β subunit of F1 to inhibit.121
Citreoviridin
This compound consists of an α-pyrone ring conjugated to a furan ring.73 It inhibits MAS by binding to the β subunit of F1, inhibiting in turn the proliferation and growth of lung adenocarcinoma cell lines. The unfolded protein response is activated without affecting healthy cells.41,77,122
Aurovertin B
This naturally occurring antibiotic is isolated from Calcarisporium arbuscular. It also interacts with F1’s β subunit and inhibits ATP synthesis.80 Aurovertin B strongly inhibits the proliferation of several breast cancer cell lines by suppressing the enhanced ATP synthesis in breast carcinoma.50 Aurovertin B, D, and E exhibit potent antiproliferative activity, particularly against triple-negative breast cancer in vivo and in vitro.123,124 They also facilitate the recognition and lysis of colon cancer cells by natural killer (NK) cells, by a mechanism that may involve MAS inhibition.81
Asteltoxin
This α-pyrone-containing mycotoxin is produced in Aspergillus stellatus Curzi and Emericella variecolor. Asteltoxin and its three dimers exert inhibitory effects on the lung cancer cell line H1299 and breast cancer line MCF7.82 Asteltoxin and asteltoxin B are moderately cytotoxic against several tumor cell lines.83
Synthetic compounds
In addition to the mentioned natural compounds, several synthetic MAS inhibitors have been developed for treating infectious diseases such as tuberculosis by inhibiting ATP synthase.125 Some were developed using computational science, with some working against cancer.5
Bedaquiline
This compound, developed by Janssen Pharmaceuticals in 2005, was approved by the US Food and Drug Administration (FDA) in 2012 for treating multidrug-resistant tuberculosis.125,126 It is a synthetic heterocyclic MAS synthase inhibitor that targets both the c-subunit rotor and subunit ε.84 Recently, Wu et al.85 suggested that bedaquiline blocks the growth, survival, and tumor angiogenesis of lung cancer, stem-like cancer cells, and glioblastoma.86, 87, 88 Similar to other ATP synthase inhibitors, it diminishes mitochondrial oxygen consumption by depleting and blocking the proliferative expansion of breast cancer stem cells.86
Gboxin
This small molecule was found by a cell-based high-throughput chemical screen. It inhibits the growth of mouse and human glioblastoma cells.5 A Pd(II) complex of a Gboxin analog-chitooligosaccharides conjugate (Pd(II)COS@GbA) was found to disrupt ATP synthase rotation, diminish ATP synthesis, and show anti-prostate cancer activity.91
Alkylphospholipid analogs (APLs) are developed as anticancer drugs, with amphiphilic structures similar to natural phospholipids, and they incorporate preferentially in the membranes of tumor cells.127 Structurally related anti-tumor APLs include a number of clinically promising drugs, such as edelfosine, miltefosine, perifosine, and erucylphosphocholine.92,96,97
Edelfosine, with antileishmanial and anticancer activity, specifically accumulated in the mitochondria or the lipid raft of tumor cells, leading to dissipation of the mitochondrial membrane potential and reactive oxygen species (ROS) generation by ATP synthase inhibition and raft disruption, eventually inducing apoptosis-like cell death.92,93
Miltefosine, also belonging to the APLs family and trademarked as Miltex, is the only APL that has been approved as an antitumor drug in the topical treatment of metastatic skin lesions in breast cancer.96
Perifosine and erucylphosphocholine are the new molecules synthesized as miltefosine variants.97 Perifosine has entered many phase I clinical trials in treating plenty of advanced solid and leukemic tumors, and many preclinical studies are being carried out with erucylphosphocholine in brain tumors.98
Peptide inhibitors
Several peptide inhibitors, including the endogenous MAS inhibitory factor 1 (IF1), angiostatin, and some chimeric antibodies, target MAS or ectopic ATP synthase, showing an anti-tumor activity in preclinical studies.
IF1
This protein interacts with the F1 domain when dephosphorylated at the catalytic surface between the α and β subunit of MAS, blocking the rotation of the complex and inhibiting the synthesis and hydrolysis of ATP.99 Its phosphorylation status is regulated by 5′-AMP-activated protein kinase (AMPK). Increased cyclic AMP (cAMP) levels activate AMPK: the phosphorylation of S39 in IF1 and the activation of MAS thus facilitate ATP synthesis.128 High expression of IF1 in non-small cell lung cancer, bladder carcinoma, and gliomas has a bad prognosis.10 A recent study demonstrated that IF1 modulates the interplay between ROS and hypoxia in cancer cells, which might be associated with the cell survival supports, tumor progression, and anticancer drug resistance.10 In contrast, a high expression level of IF1 in breast and colon cancer predicts a better prognosis, especially in triple-negative breast cancer and metastatic colorectal cancer patients.10,100 Thus, the comprehensive role of IF1 in malignancy needs subtle stratification to help anticancer therapeutic strategies.
Angiostatin
This protein is an internal proteolytic fragment of plasminogen, dramatically inhibiting primary and metastatic tumor growth by blocking tumor angiogenesis.129 EAS is relevant to angiogenesis and the direct target of angiostatin.130 Angiostatin binds the α/β subunits of EAS, mediating the downregulation of endothelial cell proliferation and migration, and these effects will be depleted by the presence of the anti-α-subunit ATP synthase antibody. Hence, inhibition of EAS has been suggested for the antiangiogenetic therapeutic strategy by blocking tumor angiogenesis.130,131 Moreover, radiolabeled angiostatin may be a powerful tool for specific imaging or targeted radiotherapy of tumors due to the specific interaction.102
Other chimeric antibodies have been developed that target the β subunit of EAS, inhibiting its activity.132 A strain of murine monoclonal antibody (mAb) mAb6F2C4 showed the angiostatin-like property to block extracellular ATP generation under extracellular acidic conditions and interrupt the angiogenetic process.14 Another murine mAb targeting the same site, McAb178-5G10, showed antitumor activity in multiple cancers.12,133 The humanized antibody Hai178 against the β subunit of EAS produced by the same team also had an anti-tumor effect in tumor xenografts,103 which provides a possibility for clinical application. This evidence ensures the potential of antibodies blocking EAS activity in cancer treatment.
Discussion
MAS is upregulated in many cancers, and evidence is accumulating that MAS may serve as a potent anticancer target, especially in glioblastoma, ovarian cancer, prostate cancer, breast cancer, and clear cell renal cell carcinoma.26, 27, 28, 29, 30,36,134,135 However, there remain several challenges in translating preclinical MAS-targeting drugs to the clinic.
First, MAS universally exists in tumor cells and normal cells, therefore raising the concern about the potential of systematic toxicity to patients. According to ClinicalTrials.gov (https://www.clinicaltrials.gov/), the potential side effects caused by various MAS inhibitors are different. There are five MAS inhibitors that have reported clinical trial results. Resveratrol was safe and lacked serious adverse reactions, with mild gastrointestinal symptoms, including nausea, flatulence, abdominal discomfort, and diarrhea (ClinicalTrials.gov: NCT00098969). EGCG was well tolerated with minimal adverse events, including nausea, diarrhea, headache. These side effects were all grade 1 or 2 based on the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 3.0 (ClinicalTrials.gov: NCT00666562). Curcumin was safe, and no adverse events occurred during the study (ClinicalTrials.gov: NCT01042938). The incidence rate of serious adverse events caused by using estradiol was 23.53%, which included abdominal pain, diarrhea, nausea, and vomiting. All patients reported mild adverse events in the study (ClinicalTrials.gov: NCT00324259). The incidence rate of serious adverse events by using genistein was 9.68%, including dyspnea and ulceration. 80.65% of the patients reported mild adverse events in this study (ClinicalTrials.gov: NCT00290758).
To overcome the shortcomings of potential toxicity, (1) antibody drug conjugates (ADCs) can be a strategy to enhance the specificity of MAS-targeting drugs. ADCs represent a new class of cancer-targeting drugs that endow the specificity to cytotoxic chemicals by conjugating them to cancer antigen-specific antibodies. Antibody-assisted drug delivery dramatically reduces the systematic toxicity to cancer patients (e.g., T-DM1). T-DM1 significantly improved overall survival of patients with previously treated HER2+ metastatic breast cancer,136 and the rates of adverse events of grade 3 or above were reduced from 57% to 41%.137 (2) VDAC1 is a voltage-dependent anion channel protein that is a major component of the outer mitochondrial membrane, often overexpressed in many cancers.138 Studies have shown that silencing VDAC1 expression in cancer cells leads to metabolic reprogramming, reducing the expression of ATP5F1A.139, 140, 141, 142 Therefore, silencing VDAC1 may decrease the level of ATP synthase without affecting healthy cells. (3) Another strategy to improve the efficacy/toxicity balance is to combine the energy supply blockers from MAS inhibitors with agents of other anti-cancer mechanisms. For instance, combination of the MAS inhibitor citreoviridin and the 26S proteasome inhibitor bortezomib improved anticancer activity in breast cancer cells.78 Combinatory use of multi-mechanism anti-cancer drugs is the basic and mostly chosen regimen for cancers at advanced stages.
Patel et al.47 provided an update on recently discovered ATP synthase modulators. A more comprehensive view of ATP synthase-targeting inhibitors was covered in their study. Because we have concentrated preferentially on the indications of ATP synthase-targeting inhibitors in cancer treatment, some natural inhibitors of the complex that have systemic toxicity or about which there is almost no relevant research in cancer treatment are not included in our review. We have elaborated the structure of the complex, detailed its significance in different types of malignant tumors, and reviewed the basic and clinical research progress of ATP synthase-targeting inhibitors in cancer treatment. A larger-scale and comprehensive screening of MAS inhibitors should be performed with the current high-throughput chemical screening technology to find more potential effective and specific anti-cancer drugs.
Second, the expression levels of the MAS-composing genes, the status of OXPHOS, and mitochondrial metabolism vary in different types of cancer.143,144 High expression of ATP5B predicts better local recurrence-free survival in nasopharyngeal carcinoma.134 In papillary thyroid cancer, ATP5A1 mRNA expression levels are downregulated in tumorous tissue with more lymph node metastases.145 Similarly, ATP5B is downregulated in breast, gastric, lung, and esophageal cancer.135 Thus, it is necessary to measure the status and inhibitory contribution of MAS before using a MAS inhibitor on tumor or in combination with other anticancer drugs.
Additionally, the challenge faced to make the MAS a dominant antitumor target should be ideally resolved. Only a certain part of tumors, which are still under to-be-defined conditions, are potentially sensitive to MAS inhibitors. For instance, mitoxantrone significantly benefits the progression-free and overall survival of cancer patients. However, in metastatic breast cancer, the response rate of mitoxantrone is only 20%.146 The expression of MAS was increased in mitoxantrone-resistant cells, so MAS might be a key target to reverse the resistance of metastatic breast cancer patients to this drug.31 Therefore, in certain cancers, the value of this complex as a target to suppress primary cancer or reverse the resistance of tumor cell to the existing treatment should be determined.
Third, MAS is mutated in some primary human cancers. A total of 19 genes are involved in encoding ATP synthase. Except for MT-ATP6 and MT-ATP8, which are mitochondrial genes, the others are nuclear genes. According to The Cancer Genome Atlas (TCGA) database, the variation frequencies of these genes in pan cancers range from 0.5% to 2%, including structure variation, amplification, deep deletion, and other point mutations. Although not many relevant results have been reported, the abnormality of MAS composing genes on the binding sites of ATP inhibitors in about 2% of tumor patients may lead to a less sensitivity to the drugs. Given that condition, we may consider the use of other inhibitors that target different sites of ATP synthase, or redesign the inhibitor to specifically bind the mutated ATP synthase, hence inhibiting the activity of the enzyme.
Fourth, metabolism reprogramming as a process that occurred in cancer cells influences not only the cancer cells but also the microenvironment.147 For instance, lipid accumulation in myeloid-derived suppressor cells and tumor-associated macrophages, which may derive from adjacent cancer cells with enhanced fatty acid synthesis, has been verified to promote metabolic reprogramming and assisted these cells changing into immunosuppressive phenotypes.148 Additionally, tumor cells consume more glucose, which results in a glucose limitation for T cells, leading to a defective antitumor response.149 Metabolite imbalance aroused by enhanced MAS in tumor cells may provide an advantageous immune microenvironment for tumor progression. Interfering with the MAS-related metabolite deregulation may be a breakthrough auxiliary approach to phenotypically renormalize the tumors to a sluggish status.
Additionally, previous studies have shown that estrogen supported the growth of tumors by activating a series of signaling pathways, and sustained exposure to exogenous estrogen was a risk factor for various cancers.150 Anti-estrogen therapies are important for cancers, especially for estrogen receptor-positive breast cancer.151 However, the function of estrogen as an ATP synthase inhibitor suppressing the activity of MAS and inducing mitochondrial functional damage and cell apoptosis have been discussed in this review. The distribution of the binding of estrogen on (1) MAS and on (2) the estrogen receptor has in general not been determined in the past, and it should be measured in future studies. This binding should also be considered and measured when using endocrine therapy on cancers such as breast cancer, ovarian cancer, and endometrial cancers.
In conclusion, MAS-targeted drugs represent an emerging family of anti-cancer agents in active development and should be identified and tested in further clinical trials to improve cancer patient outcomes in the future.
Acknowledgments
This work was supported by grants from the National Natural Science Foundation of China (nos. 81874122, 81872280, and 82073094), the Open Issue of State Key Laboratory of Molecular Oncology (no. SKL-KF-2017-16), and the Independent Issue of State Key Laboratory of Molecular Oncology (no. SKL-2017-16).
Author contributions
All listed authors contributed to the writing of this review. All authors read and provided critical revision of the manuscript and approved the final version.
Declaration of interests
The authors declare no competing interest.
Contributor Information
Fei Ma, Email: mafei2011@139.com.
Hai-li Qian, Email: qianhaili001@163.com.
References
- 1.Liberti M.V., Locasale J.W. The Warburg effect: How does it benefit cancer cells? Trends Biochem. Sci. 2016;41:211–218. doi: 10.1016/j.tibs.2015.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hanahan D., Weinberg R.A. Hallmarks of cancer: The next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 3.Pavlova N.N., Thompson C.B. The emerging hallmarks of cancer metabolism. Cell Metab. 2016;23:27–47. doi: 10.1016/j.cmet.2015.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Caro P., Kishan A.U., Norberg E., Stanley I.A., Chapuy B., Ficarro S.B., Polak K., Tondera D., Gounarides J., Yin H. Metabolic signatures uncover distinct targets in molecular subsets of diffuse large B cell lymphoma. Cancer Cell. 2012;22:547–560. doi: 10.1016/j.ccr.2012.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Shi Y., Lim S.K., Liang Q., Iyer S.V., Wang H.Y., Wang Z., Xie X., Sun D., Chen Y.J., Tabar V. Gboxin is an oxidative phosphorylation inhibitor that targets glioblastoma. Nature. 2019;567:341–346. doi: 10.1038/s41586-019-0993-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bajzikova M., Kovarova J., Coelho A.R., Boukalova S., Oh S., Rohlenova K., Svec D., Hubackova S., Endaya B., Judasova K. Reactivation of dihydroorotate dehydrogenase-driven pyrimidine biosynthesis restores tumor growth of respiration-deficient cancer cells. Cell Metab. 2019;29:399–416. doi: 10.1016/j.cmet.2018.10.014. e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Viale A., Draetta G.F. Metabolic features of cancer treatment resistance. Recent Results Cancer Res. 2016;207:135–156. doi: 10.1007/978-3-319-42118-6_6. [DOI] [PubMed] [Google Scholar]
- 8.Bosc C., Selak M.A., Sarry J.E. Resistance is futile: Targeting mitochondrial energetics and metabolism to overcome drug resistance in cancer treatment. Cell Metab. 2017;26:705–707. doi: 10.1016/j.cmet.2017.10.013. [DOI] [PubMed] [Google Scholar]
- 9.Viale A., Pettazzoni P., Lyssiotis C.A., Ying H., Sánchez N., Marchesini M., Carugo A., Green T., Seth S., Giuliani V. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature. 2014;514:628–632. doi: 10.1038/nature13611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Esparza-Moltó P.B., Cuezva J.M. The role of mitochondrial H+-ATP synthase in cancer. Front. Oncol. 2018;8:53. doi: 10.3389/fonc.2018.00053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Das B., Mondragon M.O., Sadeghian M., Hatcher V.B., Norin A.J. A novel ligand in lymphocyte-mediated cytotoxicity: Expression of the beta subunit of H+ transporting ATP synthase on the surface of tumor cell lines. J. Exp. Med. 1994;180:273–281. doi: 10.1084/jem.180.1.273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Wang W.J., Shi X.X., Liu Y.W., He Y.Q., Wang Y.Z., Yang C.X., Gao F. The mechanism underlying the effects of the cell surface ATP synthase on the regulation of intracellular acidification during acidosis. J. Cell. Biochem. 2013;114:1695–1703. doi: 10.1002/jcb.24511. [DOI] [PubMed] [Google Scholar]
- 13.Ma Z., Cao M., Liu Y., He Y., Wang Y., Yang C., Wang W., Du Y., Zhou M., Gao F. Mitochondrial F1Fo-ATP synthase translocates to cell surface in hepatocytes and has high activity in tumor-like acidic and hypoxic environment. Acta Biochim. Biophys. Sin. (Shanghai) 2010;42:530–537. doi: 10.1093/abbs/gmq063. [DOI] [PubMed] [Google Scholar]
- 14.Wang J., Han Y., Liang J., Cheng X., Yan L., Wang Y., Liu J., Luo G., Chen X., Zhao L. Effect of a novel inhibitory mAb against β-subunit of F1F0 ATPase on HCC. Cancer Biol. Ther. 2008;7:1829–1835. doi: 10.4161/cbt.7.11.6861. [DOI] [PubMed] [Google Scholar]
- 15.Chang Y.W., Hsu C.L., Tang C.W., Chen X.J., Huang H.C., Juan H.F. Multiomics reveals ectopic ATP synthase blockade induces cancer cell death via a lncRNA-mediated phospho-signaling network. Mol. Cell. Proteomics. 2020;19:1805–1825. doi: 10.1074/mcp.RA120.002219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kim M.S., Gernapudi R., Cedeno Y.C., Polster B.M., Martinez R., Shapiro P., Kesari S., Nurmemmedov E., Passaniti A. Targeting breast cancer metabolism with a novel inhibitor of mitochondrial ATP synthesis. Oncotarget. 2020;11:3863–3885. doi: 10.18632/oncotarget.27743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Okuno D., Iino R., Noji H. Rotation and structure of FoF1-ATP synthase. J. Biochem. 2011;149:655–664. doi: 10.1093/jb/mvr049. [DOI] [PubMed] [Google Scholar]
- 18.Pinke G., Zhou L., Sazanov L.A. Cryo-EM structure of the entire mammalian F-type ATP synthase. Nat. Struct. Mol. Biol. 2020;27:1077–1085. doi: 10.1038/s41594-020-0503-8. [DOI] [PubMed] [Google Scholar]
- 19.Srivastava A.P., Luo M., Zhou W., Symersky J., Bai D., Chambers M.G., Faraldo-Gomez J.D., Liao M., Mueller D.M. High-resolution cryo-EM analysis of the yeast ATP synthase in a lipid membrane. Science. 2018;360:eaas9699. doi: 10.1126/science.aas9699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Spikes T.E., Montgomery M.G., Walker J.E. Structure of the dimeric ATP synthase from bovine mitochondria. Proc. Natl. Acad. Sci. USA. 2020;117:23519–23526. doi: 10.1073/pnas.2013998117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.He J., Ford H.C., Carroll J., Douglas C., Gonzales E., Ding S., Fearnley I.M., Walker J.E. Assembly of the membrane domain of ATP synthase in human mitochondria. Proc. Natl. Acad. Sci. USA. 2018;115:2988–2993. doi: 10.1073/pnas.1722086115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Lieberthal W., Menza S.A., Levine J.S. Graded ATP depletion can cause necrosis or apoptosis of cultured mouse proximal tubular cells. Am. J. Physiol. 1998;274:F315–F327. doi: 10.1152/ajprenal.1998.274.2.F315. [DOI] [PubMed] [Google Scholar]
- 23.Niedzwiecka K., Tisi R., Penna S., Lichocka M., Plochocka D., Kucharczyk R. Two mutations in mitochondrial ATP6 gene of ATP synthase, related to human cancer, affect ROS, calcium homeostasis and mitochondrial permeability transition in yeast. Biochim. Biophys. Acta Mol. Cell. Res. 2018;1865:117–131. doi: 10.1016/j.bbamcr.2017.10.003. [DOI] [PubMed] [Google Scholar]
- 24.Li J., Agarwal E., Bertolini I., Seo J.H., Caino M.C., Ghosh J.C., Kossenkov A.V., Liu Q., Tang H.Y., Goldman A.R. The mitophagy effector FUNDC1 controls mitochondrial reprogramming and cellular plasticity in cancer cells. Sci. Signal. 2020;13:eaaz8240. doi: 10.1126/scisignal.aaz8240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Gale M., Li Y., Cao J., Liu Z.Z., Holmbeck M.A., Zhang M., Lang S.M., Wu L., Do Carmo M., Gupta S. Acquired resistance to HER2-targeted therapies creates vulnerability to ATP synthase inhibition. Cancer Res. 2020;80:524–535. doi: 10.1158/0008-5472.CAN-18-3985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Xu G., Li J.Y. ATP5A1 and ATP5B are highly expressed in glioblastoma tumor cells and endothelial cells of microvascular proliferation. J. Neurooncol. 2016;126:405–413. doi: 10.1007/s11060-015-1984-x. [DOI] [PubMed] [Google Scholar]
- 27.Muys B.R., Sousa J.F., Plaça J.R., de Araújo L.F., Sarshad A.A., Anastasakis D.G., Wang X., Li X.L., de Molfetta G.A., Ramão A. miR-450a acts as a tumor suppressor in ovarian cancer by regulating energy metabolism. Cancer Res. 2019;79:3294–3305. doi: 10.1158/0008-5472.CAN-19-0490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Speransky S., Serafini P., Caroli J., Bicciato S., Lippman M.E., Bishopric N.H. A novel RNA aptamer identifies plasma membrane ATP synthase beta subunit as an early marker and therapeutic target in aggressive cancer. Breast Cancer Res. Treat. 2019;176:271–289. doi: 10.1007/s10549-019-05174-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Bikas A., Jensen K., Patel A., Costello J., Kaltsas G., Hoperia V., Wartofsky L., Burman K., Vasko V. Mitotane induces mitochondrial membrane depolarization and apoptosis in thyroid cancer cells. Int. J. Oncol. 2019;55:7–20. doi: 10.3892/ijo.2019.4802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Yuan L., Chen L., Qian K., Wang G., Lu M., Qian G., Cao X., Jiang W., Xiao Y., Wang X. A novel correlation between ATP5A1 gene expression and progression of human clear cell renal cell carcinoma identified by co-expression analysis. Oncol. Rep. 2018;39:525–536. doi: 10.3892/or.2017.6132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chang F.W., Fan H.C., Liu J.M., Fan T.P., Jing J., Yang C.L., Hsu R.J. Estrogen enhances the expression of the multidrug transporter gene ABCG2—Increasing drug resistance of breast cancer cells through estrogen receptors. Int. J. Mol. Sci. 2017;18:E163. doi: 10.3390/ijms18010163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Feichtinger R.G., Schäfer G., Seifarth C., Mayr J.A., Kofler B., Klocker H. Reduced levels of ATP synthase subunit ATP5F1A correlate with earlier-onset prostate cancer. Oxid. Med. Cell. Longev. 2018;2018:1347174. doi: 10.1155/2018/1347174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Seth R., Keeley J., Abu-Ali G., Crook S., Jackson D., Ilyas M. The putative tumour modifier gene ATP5A1 is not mutated in human colorectal cancer cell lines but expression levels correlate with TP53 mutations and chromosomal instability. J. Clin. Pathol. 2009;62:598–603. doi: 10.1136/jcp.2009.064436. [DOI] [PubMed] [Google Scholar]
- 34.Liu T., Guo J., Wang T., Zhang S., Yu X., Hou C., Guo D. Network pharmacology-based analysis of mechanisms of the anti-hepatocellular carcinoma effect by dihydroartemisinin. Discov. Med. 2019;28:139–147. [PubMed] [Google Scholar]
- 35.Lu Z., Song Q., Yang J., Zhao X., Zhang X., Yang P., Kang J. Comparative proteomic analysis of anti-cancer mechanism by periplocin treatment in lung cancer cells. Cell. Physiol. Biochem. 2014;33:859–868. doi: 10.1159/000358658. [DOI] [PubMed] [Google Scholar]
- 36.Ratajczak K., Lukasiak A., Grel H., Dworakowska B., Jakiela S., Stobiecka M. Monitoring of dynamic ATP level changes by oligomycin-modulated ATP synthase inhibition in SW480 cancer cells using fluorescent “On-Off” switching DNA aptamer. Anal. Bioanal. Chem. 2019;411:6899–6911. doi: 10.1007/s00216-019-02061-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Di Virgilio F., Sarti A.C., Falzoni S., De Marchi E., Adinolfi E. Extracellular ATP and P2 purinergic signalling in the tumour microenvironment. Nat. Rev. Cancer. 2018;18:601–618. doi: 10.1038/s41568-018-0037-0. [DOI] [PubMed] [Google Scholar]
- 38.Pellegatti P., Raffaghello L., Bianchi G., Piccardi F., Pistoia V., Di Virgilio F. Increased level of extracellular ATP at tumor sites: In vivo imaging with plasma membrane luciferase. PLoS ONE. 2008;3:e2599. doi: 10.1371/journal.pone.0002599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Astakhova A., Chistyakov D., Thomas D., Geisslinger G., Brüne B., Sergeeva M., Namgaladze D. Inhibitors of oxidative phosphorylation modulate astrocyte inflammatory responses through AMPK-dependent Ptgs2 mRNA stabilization. Cells. 2019;8:E1185. doi: 10.3390/cells8101185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Park B.N., Kim G.H., Ko S.A., Shin G.H., Lee S.J., An Y.S., Yoon J.K. Zr-89 immuno-PET targeting ectopic ATP synthase enables in-vivo imaging of tumor angiogenesis. Int. J. Mol. Sci. 2019;20:E3928. doi: 10.3390/ijms20163928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chang H.Y., Huang H.C., Huang T.C., Yang P.C., Wang Y.C., Juan H.F. Ectopic ATP synthase blockade suppresses lung adenocarcinoma growth by activating the unfolded protein response. Cancer Res. 2012;72:4696–4706. doi: 10.1158/0008-5472.CAN-12-0567. [DOI] [PubMed] [Google Scholar]
- 42.Wang T., Shen Y., Li Y., Wang B., Wang B., Wu D., Ruan C., Wang Y. Ectopic ATP synthase β subunit proteins on human leukemia cell surface interact with platelets by binding glycoprotein IIb. Haematologica. 2019;104:e364–e368. doi: 10.3324/haematol.2019.216390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bianchi G., Ravera S., Traverso C., Amaro A., Piaggio F., Emionite L., Bachetti T., Pfeffer U., Raffaghello L. Curcumin induces a fatal energetic impairment in tumor cells in vitro and in vivo by inhibiting ATP-synthase activity. Carcinogenesis. 2018;39:1141–1150. doi: 10.1093/carcin/bgy076. [DOI] [PubMed] [Google Scholar]
- 44.Salomon A.R., Voehringer D.W., Herzenberg L.A., Khosla C. Apoptolidin, a selective cytotoxic agent, is an inhibitor of F0F1-ATPase. Chem. Biol. 2001;8:71–80. doi: 10.1016/s1074-5521(00)00057-0. [DOI] [PubMed] [Google Scholar]
- 45.Mowery Y.M., Pizzo S.V. Targeting cell surface F1F0 ATP synthase in cancer therapy. Cancer Biol. Ther. 2008;7:1836–1838. doi: 10.4161/cbt.7.11.7155. [DOI] [PubMed] [Google Scholar]
- 46.Hong S., Pedersen P.L. ATP synthase and the actions of inhibitors utilized to study its roles in human health, disease, and other scientific areas. Microbiol. Mol. Biol. Rev. 2008;72:590–641. doi: 10.1128/MMBR.00016-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Patel B.A., D’Amico T.L., Blagg B. Natural products and other inhibitors of F1FO ATP synthase. Eur. J. Med. Chem. 2020;207:112779. doi: 10.1016/j.ejmech.2020.112779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Narang R., Kumar R., Kalra S., Nayak S.K., Khatik G.L., Kumar G.N., Sudhakar K., Singh S.K. Recent advancements in mechanistic studies and structure activity relationship of FoF1 ATP synthase inhibitor as antimicrobial agent. Eur. J. Med. Chem. 2019;182:111644. doi: 10.1016/j.ejmech.2019.111644. [DOI] [PubMed] [Google Scholar]
- 49.Arakaki N., Kita T., Shibata H., Higuti T. Cell-surface H+-ATP synthase as a potential molecular target for anti-obesity drugs. FEBS Lett. 2007;581:3405–3409. doi: 10.1016/j.febslet.2007.06.041. [DOI] [PubMed] [Google Scholar]
- 50.Huang T.C., Chang H.Y., Hsu C.H., Kuo W.H., Chang K.J., Juan H.F. Targeting therapy for breast carcinoma by ATP synthase inhibitor aurovertin B. J. Proteome Res. 2008;7:1433–1444. doi: 10.1021/pr700742h. [DOI] [PubMed] [Google Scholar]
- 51.Devenish R.J., Prescott M., Boyle G.M., Nagley P. The oligomycin axis of mitochondrial ATP synthase: OSCP and the proton channel. J. Bioenerg. Biomembr. 2000;32:507–515. doi: 10.1023/a:1005621125812. [DOI] [PubMed] [Google Scholar]
- 52.Tan J.L., Li F., Yeo J.Z., Yong K.J., Bassal M.A., Ng G.H., Lee M.Y., Leong C.Y., Tan H.K., Wu C.S. New high-throughput screening identifies compounds that reduce viability specifically in liver cancer cells that express high levels of SALL4 by inhibiting oxidative phosphorylation. Gastroenterology. 2019;157:1615–1629.e17. doi: 10.1053/j.gastro.2019.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Lin C.S., Liu L.T., Ou L.H., Pan S.C., Lin C.I., Wei Y.H. Role of mitochondrial function in the invasiveness of human colon cancer cells. Oncol. Rep. 2018;39:316–330. doi: 10.3892/or.2017.6087. [DOI] [PubMed] [Google Scholar]
- 54.Wender P.A., Longcore K.E. Isolation, structure determination, and anti-cancer activity of apoptolidin D. Org. Lett. 2007;9:691–694. doi: 10.1021/ol0630245. [DOI] [PubMed] [Google Scholar]
- 55.Wender P.A., Sukopp M., Longcore K. Apoptolidins B and C: Isolation, structure determination, and biological activity. Org. Lett. 2005;7:3025–3028. doi: 10.1021/ol051074o. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wang S., Moustaid-Moussa N., Chen L., Mo H., Shastri A., Su R., Bapat P., Kwun I., Shen C.L. Novel insights of dietary polyphenols and obesity. J. Nutr. Biochem. 2014;25:1–18. doi: 10.1016/j.jnutbio.2013.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Rauf A., Imran M., Butt M.S., Nadeem M., Peters D.G., Mubarak M.S. Resveratrol as an anti-cancer agent: A review. Crit. Rev. Food Sci. Nutr. 2018;58:1428–1447. doi: 10.1080/10408398.2016.1263597. [DOI] [PubMed] [Google Scholar]
- 58.Fliedner S.M., Yang C., Thompson E., Abu-Asab M., Hsu C.M., Lampert G., Eiden L., Tischler A.S., Wesley R., Zhuang Z. Potential therapeutic target for malignant paragangliomas: ATP synthase on the surface of paraganglioma cells. Am. J. Cancer Res. 2015;5:1558–1570. [PMC free article] [PubMed] [Google Scholar]
- 59.Valenti D., de Bari L., Manente G.A., Rossi L., Mutti L., Moro L., Vacca R.A. Negative modulation of mitochondrial oxidative phosphorylation by epigallocatechin-3 gallate leads to growth arrest and apoptosis in human malignant pleural mesothelioma cells. Biochim. Biophys. Acta. 2013;1832:2085–2096. doi: 10.1016/j.bbadis.2013.07.014. [DOI] [PubMed] [Google Scholar]
- 60.Khiewkamrop P., Phunsomboon P., Richert L., Pekthong D., Srisawang P. Epistructured catechins, EGCG and EC facilitate apoptosis induction through targeting de novo lipogenesis pathway in HepG2 cells. Cancer Cell Int. 2018;18:46. doi: 10.1186/s12935-018-0539-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Liu M., Xu Y.F., Feng Y., Yang F.Q., Luo J., Zhai W., Che J.P., Wang G.C., Zheng J.H. Epigallocatechin gallate attenuates interstitial cystitis in human bladder urothelium cells by modulating purinergic receptors. J. Surg. Res. 2013;183:397–404. doi: 10.1016/j.jss.2012.11.041. [DOI] [PubMed] [Google Scholar]
- 62.Nelson K.M., Dahlin J.L., Bisson J., Graham J., Pauli G.F., Walters M.A. The essential medicinal chemistry of curcumin. J. Med. Chem. 2017;60:1620–1637. doi: 10.1021/acs.jmedchem.6b00975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Rawat D., Shrivastava S., Naik R.A., Chhonker S.K., Mehrotra A., Koiri R.K. An overview of natural plant products in the treatment of hepatocellular carcinoma. Anticancer. Agents Med. Chem. 2018;18:1838–1859. doi: 10.2174/1871520618666180604085612. [DOI] [PubMed] [Google Scholar]
- 64.Dang Y.P., Yuan X.Y., Tian R., Li D.G., Liu W. Curcumin improves the paclitaxel-induced apoptosis of HPV-positive human cervical cancer cells via the NF-κB-p53-caspase-3 pathway. Exp. Ther. Med. 2015;9:1470–1476. doi: 10.3892/etm.2015.2240. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 65.Faião-Flores F., Suarez J.A., Pardi P.C., Maria D.A. DM-1, sodium 4-[5-(4-hydroxy-3-methoxyphenyl)-3-oxo-penta-1,4-dienyl]-2-methoxy-phenolate: A curcumin analog with a synergic effect in combination with paclitaxel in breast cancer treatment. Tumour Biol. 2012;33:775–785. doi: 10.1007/s13277-011-0293-z. [DOI] [PubMed] [Google Scholar]
- 66.Chen Q.H. Curcumin-based anti-prostate cancer agents. Anticancer. Agents Med. Chem. 2015;15:138–156. doi: 10.2174/1871520615666150116102442. [DOI] [PubMed] [Google Scholar]
- 67.Wan Mohd Tajuddin W.N.B., Lajis N.H., Abas F., Othman I., Naidu R. Mechanistic understanding of curcumin’s therapeutic effects in lung cancer. Nutrients. 2019;11:E2989. doi: 10.3390/nu11122989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Deguchi A. Curcumin targets in inflammation and cancer. Endocr. Metab. Immune Disord. Drug Targets. 2015;15:88–96. doi: 10.2174/1871530315666150316120458. [DOI] [PubMed] [Google Scholar]
- 69.Alvarez-Delgado C., Mendoza-Rodriguez C.A., Picazo O., Cerbon M. Different expression of alpha and beta mitochondrial estrogen receptors in the aging rat brain: Interaction with respiratory complex V. Exp. Gerontol. 2010;45:580–585. doi: 10.1016/j.exger.2010.01.015. [DOI] [PubMed] [Google Scholar]
- 70.Massart F., Paolini S., Piscitelli E., Brandi M.L., Solaini G. Dose-dependent inhibition of mitochondrial ATP synthase by 17 beta-estradiol. Gynecol. Endocrinol. 2002;16:373–377. [PubMed] [Google Scholar]
- 71.Zheng J., Ramirez V.D. Purification and identification of an estrogen binding protein from rat brain: Oligomycin sensitivity-conferring protein (OSCP), a subunit of mitochondrial F0F1-ATP synthase/ATPase. J. Steroid Biochem. Mol. Biol. 1999;68:65–75. doi: 10.1016/s0960-0760(98)00161-7. [DOI] [PubMed] [Google Scholar]
- 72.Zheng J., Ramirez V.D. Rapid inhibition of rat brain mitochondrial proton F0F1-ATPase activity by estrogens: comparison with Na+, K+-ATPase of porcine cortex. Eur. J. Pharmacol. 1999;368:95–102. doi: 10.1016/s0014-2999(99)00012-6. [DOI] [PubMed] [Google Scholar]
- 73.Pitt J.I., Miller J.D. A concise history of mycotoxin research. J. Agric. Food Chem. 2017;65:7021–7033. doi: 10.1021/acs.jafc.6b04494. [DOI] [PubMed] [Google Scholar]
- 74.Kumar A., Banerjee A., Singh D., Thakur G., Kasarpalkar N., Gavali S., Gadkar S., Madan T., Mahale S.D., Balasinor N.H., Sachdeva G. Estradiol: A steroid with multiple facets. Horm. Metab. Res. 2018;50:359–374. doi: 10.1055/s-0044-100920. [DOI] [PubMed] [Google Scholar]
- 75.Chinnam N., Dadi P.K., Sabri S.A., Ahmad M., Kabir M.A., Ahmad Z. Dietary bioflavonoids inhibit Escherichia coli ATP synthase in a differential manner. Int. J. Biol. Macromol. 2010;46:478–486. doi: 10.1016/j.ijbiomac.2010.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Pons D.G., Nadal-Serrano M., Blanquer-Rossello M.M., Sastre-Serra J., Oliver J., Roca P. Genistein modulates proliferation and mitochondrial functionality in breast cancer cells depending on ERalpha/ERbeta ratio. J. Cell. Biochem. 2014;115:949–958. doi: 10.1002/jcb.24737. [DOI] [PubMed] [Google Scholar]
- 77.Gause E.M., Buck M.A., Douglas M.G. Binding of citreoviridin to the β subunit of the yeast F1-ATPase. J. Biol. Chem. 1981;256:557–559. [PubMed] [Google Scholar]
- 78.Chang H.Y., Huang T.C., Chen N.N., Huang H.C., Juan H.F. Combination therapy targeting ectopic ATP synthase and 26S proteasome induces ER stress in breast cancer cells. Cell Death Dis. 2014;5:e1540. doi: 10.1038/cddis.2014.504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Pohland A.E. Mycotoxins in review. Food Addit. Contam. 1993;10:17–28. doi: 10.1080/02652039309374126. [DOI] [PubMed] [Google Scholar]
- 80.Matsuno-Yagi A., Yagi T., Hatefi Y. Studies on the mechanism of oxidative phosphorylation: Effects of specific F0 modifiers on ligand-induced conformation changes of F1. Proc. Natl. Acad. Sci. USA. 1985;82:7550–7554. doi: 10.1073/pnas.82.22.7550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Zhu H., Wang F., Ju X., Kong L., An T., Zhao Z., Liu J., Li Y. Aurovertin B sensitizes colorectal cancer cells to NK cell recognition and lysis. Biochem. Biophys. Res. Commun. 2018;503:3057–3063. doi: 10.1016/j.bbrc.2018.08.093. [DOI] [PubMed] [Google Scholar]
- 82.Long H., Cheng Z., Huang W., Wu Q., Li X., Cui J., Proksch P., Lin W. Diasteltoxins A–C, asteltoxin-based dimers from a mutant of the sponge-associated Emericella variecolor fungus. Org. Lett. 2016;18:4678–4681. doi: 10.1021/acs.orglett.6b02313. [DOI] [PubMed] [Google Scholar]
- 83.Wu Q., Long H.L., Liu D., Proksch P., Lin W.H. Varioxiranols I-L, new lactones from a sponge-associated Emericella variecolor fungus. J. Asian Nat. Prod. Res. 2015;17:1137–1145. doi: 10.1080/10286020.2015.1119127. [DOI] [PubMed] [Google Scholar]
- 84.Preiss L., Langer J.D., Yildiz Ö., Eckhardt-Strelau L., Guillemont J.E., Koul A., Meier T. Structure of the mycobacterial ATP synthase Fo rotor ring in complex with the anti-TB drug bedaquiline. Sci. Adv. 2015;1:e1500106. doi: 10.1126/sciadv.1500106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Wu X., Li F., Wang X., Li C., Meng Q., Wang C., Huang J., Chen S., Zhu Z. Antibiotic bedaquiline effectively targets growth, survival and tumor angiogenesis of lung cancer through suppressing energy metabolism. Biochem. Biophys. Res. Commun. 2018;495:267–272. doi: 10.1016/j.bbrc.2017.10.136. [DOI] [PubMed] [Google Scholar]
- 86.Fiorillo M., Lamb R., Tanowitz H.B., Cappello A.R., Martinez-Outschoorn U.E., Sotgia F., Lisanti M.P. Bedaquiline, an FDA-approved antibiotic, inhibits mitochondrial function and potently blocks the proliferative expansion of stem-like cancer cells (CSCs) Aging (Albany NY) 2016;8:1593–1607. doi: 10.18632/aging.100983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Liu Y., Fang S., Sun Q., Liu B. Anthelmintic drug ivermectin inhibits angiogenesis, growth and survival of glioblastoma through inducing mitochondrial dysfunction and oxidative stress. Biochem. Biophys. Res. Commun. 2016;480:415–421. doi: 10.1016/j.bbrc.2016.10.064. [DOI] [PubMed] [Google Scholar]
- 88.Skoda J., Borankova K., Jansson P.J., Huang M.L., Veselska R., Richardson D.R. Pharmacological targeting of mitochondria in cancer stem cells: An ancient organelle at the crossroad of novel anti-cancer therapies. Pharmacol. Res. 2019;139:298–313. doi: 10.1016/j.phrs.2018.11.020. [DOI] [PubMed] [Google Scholar]
- 89.Cox E., Laessig K. FDA approval of bedaquiline—The benefit-risk balance for drug-resistant tuberculosis. N. Engl. J. Med. 2014;371:689–691. doi: 10.1056/NEJMp1314385. [DOI] [PubMed] [Google Scholar]
- 90.Hards K., Cook G.M. Targeting bacterial energetics to produce new antimicrobials. Drug Resist. Updat. 2018;36:1–12. doi: 10.1016/j.drup.2017.11.001. [DOI] [PubMed] [Google Scholar]
- 91.Elbehairi S.E.I., Alfaifi M.Y., Shati A.A., Alshehri M.A., Elshaarawy R.F.M., Hafez H.S. Role of Pd(II)-chitooligosaccharides-Gboxin analog in oxidative phosphorylation inhibition and energy depletion: Targeting mitochondrial dynamics. Chem. Biol. Drug Des. 2020;96:1148–1161. doi: 10.1111/cbdd.13703. [DOI] [PubMed] [Google Scholar]
- 92.Gajate C., Mollinedo F. Lipid rafts, endoplasmic reticulum and mitochondria in the antitumor action of the alkylphospholipid analog edelfosine. Anticancer. Agents Med. Chem. 2014;14:509–527. doi: 10.2174/1871520614666140309222259. [DOI] [PubMed] [Google Scholar]
- 93.Villa-Pulgarín J.A., Gajate C., Botet J., Jimenez A., Justies N., Varela-M R.E., Cuesta-Marbán Á., Müller I., Modolell M., Revuelta J.L., Mollinedo F. Mitochondria and lipid raft-located FOF1-ATP synthase as major therapeutic targets in the antileishmanial and anticancer activities of ether lipid edelfosine. PLoS Negl. Trop. Dis. 2017;11:e0005805. doi: 10.1371/journal.pntd.0005805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.van Blitterswijk W.J., Verheij M. Anticancer mechanisms and clinical application of alkylphospholipids. Biochim. Biophys. Acta. 2013;1831:663–674. doi: 10.1016/j.bbalip.2012.10.008. [DOI] [PubMed] [Google Scholar]
- 95.Mollinedo F., Fernández M., Hornillos V., Delgado J., Amat-Guerri F., Acuña A.U., Nieto-Miguel T., Villa-Pulgarín J.A., González-García C., Ceña V., Gajate C. Involvement of lipid rafts in the localization and dysfunction effect of the antitumor ether phospholipid edelfosine in mitochondria. Cell Death Dis. 2011;2:e158. doi: 10.1038/cddis.2011.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Dorlo T.P., Balasegaram M., Beijnen J.H., de Vries P.J. Miltefosine: A review of its pharmacology and therapeutic efficacy in the treatment of leishmaniasis. J. Antimicrob. Chemother. 2012;67:2576–2597. doi: 10.1093/jac/dks275. [DOI] [PubMed] [Google Scholar]
- 97.Mollinedo F. Editorial: Antitumor alkylphospholipid analogs: A promising and growing family of synthetic cell membrane-targeting molecules for cancer treatment) Anticancer. Agents Med. Chem. 2014;14:495–498. doi: 10.2174/1871520614999140313160011. [DOI] [PubMed] [Google Scholar]
- 98.Zaremberg V., Ganesan S., Mahadeo M. Lipids and membrane microdomains: The glycerolipid and alkylphosphocholine class of cancer chemotherapeutic drugs. Handb. Exp. Pharmacol. 2020;259:261–288. doi: 10.1007/164_2019_222. [DOI] [PubMed] [Google Scholar]
- 99.Esparza-Moltó P.B., Nuevo-Tapioles C., Cuezva J.M. Regulation of the H+-ATP synthase by IF1: A role in mitohormesis. Cell. Mol. Life Sci. 2017;74:2151–2166. doi: 10.1007/s00018-017-2462-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.González-Llorente L., Santacatterina F., García-Aguilar A., Nuevo-Tapioles C., González-García S., Tirpakova Z., Toribio M.L., Cuezva J.M. Overexpression of mitochondrial IF1 prevents metastatic disease of colorectal cancer by enhancing anoikis and tumor infiltration of NK cells. Cancers (Basel) 2019;12:E22. doi: 10.3390/cancers12010022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Sgarbi G., Gorini G., Liuzzi F., Solaini G., Baracca A. Hypoxia and IF1 expression promote ROS decrease in cancer cells. Cells. 2018;7:E64. doi: 10.3390/cells7070064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Jung K.H., Song S.H., Paik J.Y., Koh B.H., Choe Y.S., Lee E.J., Kim B.T., Lee K.H. Direct targeting of tumor cell F1F0 ATP-synthase by radioiodine angiostatin in vitro and in vivo. Cancer Biother. Radiopharm. 2007;22:704–712. doi: 10.1089/cbr.2007.369. [DOI] [PubMed] [Google Scholar]
- 103.Chen C., Liang H., Liao X., Pan J., Chen J., Zhao S., Xu Y., Wu Y., Ni J. A humanized chimeric antibody Hai178 targeted to the β subunit of F1F0 ATP synthase. Tumour Biol. 2016;37:15903–15912. doi: 10.1007/s13277-016-5423-1. [DOI] [PubMed] [Google Scholar]
- 104.Ridley C.P., Khosla C. In: Third Edition. Schaechter M., editor. Academic Press; 2009. Polyketides; pp. 472–481. (Encyclopedia of Microbiology). [Google Scholar]
- 105.Penefsky H.S. Mechanism of inhibition of mitochondrial adenosine triphosphatase by dicyclohexylcarbodiimide and oligomycin: Relationship to ATP synthesis. Proc. Natl. Acad. Sci. USA. 1985;82:1589–1593. doi: 10.1073/pnas.82.6.1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Giorgio V., Fogolari F., Lippe G., Bernardi P. OSCP subunit of mitochondrial ATP synthase: Role in regulation of enzyme function and of its transition to a pore. Br. J. Pharmacol. 2019;176:4247–4257. doi: 10.1111/bph.14513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Pacheco-Velázquez S.C., Robledo-Cadena D.X., Hernández-Reséndiz I., Gallardo-Pérez J.C., Moreno-Sánchez R., Rodríguez-Enríquez S. Energy metabolism drugs block triple negative breast metastatic cancer cell phenotype. Mol. Pharm. 2018;15:2151–2164. doi: 10.1021/acs.molpharmaceut.8b00015. [DOI] [PubMed] [Google Scholar]
- 108.Sun X., Zhan L., Chen Y., Wang G., He L., Wang Q., Zhou F., Yang F., Wu J., Wu Y. Increased mtDNA copy number promotes cancer progression by enhancing mitochondrial oxidative phosphorylation in microsatellite-stable colorectal cancer. Signal Transduct. Target. Ther. 2018;3:8. doi: 10.1038/s41392-018-0011-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Zhang Y., Nguyen T.T.T., Shang E., Mela A., Humala N., Mahajan A., Zhao J., Shu C., Torrini C., Sanchez-Quintero M.J. MET inhibition elicits PGC1α-dependent metabolic reprogramming in glioblastoma. Cancer Res. 2020;80:30–43. doi: 10.1158/0008-5472.CAN-19-1389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Huang Z., Kondoh E., Visco Z.R., Baba T., Matsumura N., Dolan E., Whitaker R.S., Konishi I., Fujii S., Berchuck A., Murphy S.K. Targeting dormant ovarian cancer cells in vitro and in an in vivo mouse model of platinum resistance. Mol. Cancer Ther. 2021;20:85–95. doi: 10.1158/1535-7163.MCT-20-0119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Salomon A.R., Voehringer D.W., Herzenberg L.A., Khosla C. Understanding and exploiting the mechanistic basis for selectivity of polyketide inhibitors of F0F1-ATPase. Proc. Natl. Acad. Sci. USA. 2000;97:14766–14771. doi: 10.1073/pnas.97.26.14766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Daniel P.T., Koert U., Schuppan J. Apoptolidin: Induction of apoptosis by a natural product. Angew. Chem. Int. Ed. Engl. 2006;45:872–893. doi: 10.1002/anie.200502698. [DOI] [PubMed] [Google Scholar]
- 113.Wender P.A., Jankowski O.D., Longcore K., Tabet E.A., Seto H., Tomikawa T. Correlation of F0F1-ATPase inhibition and antiproliferative activity of apoptolidin analogues. Org. Lett. 2006;8:589–592. doi: 10.1021/ol052800q. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Serrill J.D., Tan M., Fotso S., Sikorska J., Kasanah N., Hau A.M., McPhail K.L., Santosa D.A., Zabriskie T.M., Mahmud T. Apoptolidins A and C activate AMPK in metabolically sensitive cell types and are mechanistically distinct from oligomycin A. Biochem. Pharmacol. 2015;93:251–265. doi: 10.1016/j.bcp.2014.11.015. [DOI] [PubMed] [Google Scholar]
- 115.Yamashita N., Kazuo S.Y., Kitamura M., Wakao H., Furihata K., Furihata K., Hayakawa Y., Miyajima A., Seto H. Cytovaricin B, a new inhibitor of JAK-STAT signal transduction produced by Streptomyces torulosus. J. Antibiot. (Tokyo) 1997;50:440–442. doi: 10.7164/antibiotics.50.440. [DOI] [PubMed] [Google Scholar]
- 116.Estrela J.M., Mena S., Obrador E., Benlloch M., Castellano G., Salvador R., Dellinger R.W. Polyphenolic phytochemicals in cancer prevention and therapy: Bioavailability versus bioefficacy. J. Med. Chem. 2017;60:9413–9436. doi: 10.1021/acs.jmedchem.6b01026. [DOI] [PubMed] [Google Scholar]
- 117.Genestie C., Leary A., Devouassoux M., Auguste A. [Histological and molecular classification of endometrial carcinoma and therapeutical implications] Bull. Cancer. 2017;104:1001–1012. doi: 10.1016/j.bulcan.2017.08.004. [DOI] [PubMed] [Google Scholar]
- 118.Weinreb O., Amit T., Youdim M.B. The application of proteomics for studying the neurorescue activity of the polyphenol (−)-epigallocatechin-3-gallate. Arch. Biochem. Biophys. 2008;476:152–160. doi: 10.1016/j.abb.2008.01.004. [DOI] [PubMed] [Google Scholar]
- 119.Kunnumakkara A.B., Bordoloi D., Harsha C., Banik K., Gupta S.C., Aggarwal B.B. Curcumin mediates anticancer effects by modulating multiple cell signaling pathways. Clin. Sci. (Lond.) 2017;131:1781–1799. doi: 10.1042/CS20160935. [DOI] [PubMed] [Google Scholar]
- 120.Moreno A.J., Moreira P.I., Custódio J.B., Santos M.S. Mechanism of inhibition of mitochondrial ATP synthase by 17β-estradiol. J. Bioenerg. Biomembr. 2013;45:261–270. doi: 10.1007/s10863-012-9497-1. [DOI] [PubMed] [Google Scholar]
- 121.Schäberle T.F. Biosynthesis of α-pyrones. Beilstein J. Org. Chem. 2016;12:571–588. doi: 10.3762/bjoc.12.56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Hu C.W., Hsu C.L., Wang Y.C., Ishihama Y., Ku W.C., Huang H.C., Juan H.F. Temporal phosphoproteome dynamics induced by an ATP synthase inhibitor citreoviridin. Mol. Cell. Proteomics. 2015;14:3284–3298. doi: 10.1074/mcp.M115.051383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Zhao H., Wu R., Ma L.-F., Wo L.-K., Hu Y.-Y., Chen C., Zhan Z.-J. Aurovertin-type polyketides from Calcarisporium arbuscula with potent cytotoxic activities against triple-negative breast cancer. Helvetica Chimica Acta. 2016;99:543–546. [Google Scholar]
- 124.Wu R., Yang X., Zhou Q., Yu W., Li M., Wo J., Shan W., Zhao H., Chen Y., Zhan Z. Aurovertin B exerts potent antitumor activity against triple-negative breast cancer in vivo and in vitro via regulating ATP synthase activity and DUSP1 expression. Pharmazie. 2020;75:261–265. doi: 10.1691/ph.2020.0380. [DOI] [PubMed] [Google Scholar]
- 125.Calvert M.B., Furkert D.P., Cooper C.B., Brimble M.A. Synthetic approaches towards bedaquiline and its derivatives. Bioorg. Med. Chem. Lett. 2020;30:127172. doi: 10.1016/j.bmcl.2020.127172. [DOI] [PubMed] [Google Scholar]
- 126.Nieto Ramirez L.M., Quintero Vargas K., Diaz G. Whole genome sequencing for the analysis of drug resistant strains of Mycobacterium tuberculosis: A systematic review for bedaquiline and delamanid. Antibiotics (Basel) 2020;9:E133. doi: 10.3390/antibiotics9030133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Rios-Marco P., Marco C., Galvez X., Jimenez-Lopez J.M., Carrasco M.P. Alkylphospholipids: An update on molecular mechanisms and clinical relevance. Biochim. Biophys. Acta Biomembr. 2017;1859:1657–1667. doi: 10.1016/j.bbamem.2017.02.016. [DOI] [PubMed] [Google Scholar]
- 128.Garcia-Bermudez J., Sanchez-Arago M., Soldevilla B., Del A.A., Nuevo-Tapioles C., Cuezva J.M. PKA phosphorylates the ATPase inhibitory factor 1 and inactivates its capacity to bind and inhibit the mitochondrial H+-ATP synthase. Cell. Rep. 2015;12:2143–2155. doi: 10.1016/j.celrep.2015.08.052. [DOI] [PubMed] [Google Scholar]
- 129.Cao Y., Xue L. Angiostatin. Semin. Thromb. Hemost. 2004;30:83–93. doi: 10.1055/s-2004-822973. [DOI] [PubMed] [Google Scholar]
- 130.Kenan D.J., Wahl M.L. Ectopic localization of mitochondrial ATP synthase: A target for anti-angiogenesis intervention? J. Bioenerg. Biomembr. 2005;37:461–465. doi: 10.1007/s10863-005-9492-x. [DOI] [PubMed] [Google Scholar]
- 131.Moser T.L., Stack M.S., Asplin I., Enghild J.J., Højrup P., Everitt L., Hubchak S., Schnaper H.W., Pizzo S.V. Angiostatin binds ATP synthase on the surface of human endothelial cells. Proc. Natl. Acad. Sci. USA. 1999;96:2811–2816. doi: 10.1073/pnas.96.6.2811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Yuan J., Zhang C., Fang S., Zhuang Z., Ling S., Wang S. A monoclonal antibody against F1-F0 ATP synthase beta subunit. Hybridoma (Larchmt.) 2012;31:352–357. doi: 10.1089/hyb.2012.0033. [DOI] [PubMed] [Google Scholar]
- 133.Wen-Li Z., Jian W., Yan-Fang T., Xing F., Yan-Hong L., Xue-Ming Z., Min Z., Jian N., Jian P. Inhibition of the ecto-beta subunit of F1F0-ATPase inhibits proliferation and induces apoptosis in acute myeloid leukemia cell lines. J. Exp. Clin. Cancer Res. 2012;31:92. doi: 10.1186/1756-9966-31-92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Chung I.C., Chen L.C., Tsang N.M., Chuang W.Y., Liao T.C., Yuan S.N., OuYang C.N., Ojcius D.M., Wu C.C., Chang Y.S. Mitochondrial oxidative phosphorylation complex regulates NLRP3 inflammasome activation and predicts patient survival in nasopharyngeal carcinoma. Mol. Cell. Proteomics. 2020;19:142–154. doi: 10.1074/mcp.RA119.001808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Isidoro A., Martínez M., Fernández P.L., Ortega A.D., Santamaría G., Chamorro M., Reed J.C., Cuezva J.M. Alteration of the bioenergetic phenotype of mitochondria is a hallmark of breast, gastric, lung and oesophageal cancer. Biochem. J. 2004;378:17–20. doi: 10.1042/BJ20031541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Dieras V., Miles D., Verma S., Pegram M., Welslau M., Baselga J., Krop I.E., Blackwell K., Hoersch S., Xu J. Trastuzumab emtansine versus capecitabine plus lapatinib in patients with previously treated HER2-positive advanced breast cancer (EMILIA): a descriptive analysis of final overall survival results from a randomised, open-label, phase 3 trial. Lancet Oncol. 2017;18:732–742. doi: 10.1016/S1470-2045(17)30312-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Verma S., Miles D., Gianni L., Krop I.E., Welslau M., Baselga J., Pegram M., Oh D.Y., Diéras V., Guardino E., EMILIA Study Group Trastuzumab emtansine for HER2-positive advanced breast cancer. N. Engl. J. Med. 2012;367:1783–1791. doi: 10.1056/NEJMoa1209124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Arif T., Paul A., Krelin Y., Shteinfer-Kuzmine A., Shoshan-Barmatz V. Mitochondrial VDAC1 silencing leads to metabolic rewiring and the reprogramming of tumour cells into advanced differentiated states. Cancers (Basel) 2018;10:E499. doi: 10.3390/cancers10120499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Arif T., Amsalem Z., Shoshan-Barmatz V. Metabolic reprograming via silencing of mitochondrial VDAC1 expression encourages differentiation of cancer cells. Mol. Ther. Nucleic Acids. 2019;17:24–37. doi: 10.1016/j.omtn.2019.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Arif T., Stern O., Pittala S., Chalifa-Caspi V., Shoshan-Barmatz V. Rewiring of cancer cell metabolism by mitochondrial VDAC1 depletion results in time-dependent tumor reprogramming: Glioblastoma as a proof of concept. Cells. 2019;8:E1330. doi: 10.3390/cells8111330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Amsalem Z., Arif T., Shteinfer-Kuzmine A., Chalifa-Caspi V., Shoshan-Barmatz V. The mitochondrial protein VDAC1 at the crossroads of cancer cell metabolism: the epigenetic link. Cancers (Basel) 2020;12:E1031. doi: 10.3390/cancers12041031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Zerbib E., Arif T., Shteinfer-Kuzmine A., Chalifa-Caspi V., Shoshan-Barmatz V. VDAC1 silencing in cancer cells leads to metabolic reprogramming that modulates tumor microenvironment. Cancers (Basel) 2021;13:2850. doi: 10.3390/cancers13112850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Vyas S., Zaganjor E., Haigis M.C. Mitochondria and cancer. Cell. 2016;166:555–566. doi: 10.1016/j.cell.2016.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Weinberg F., Hamanaka R., Wheaton W.W., Weinberg S., Joseph J., Lopez M., Kalyanaraman B., Mutlu G.M., Budinger G.R., Chandel N.S. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc. Natl. Acad. Sci. USA. 2010;107:8788–8793. doi: 10.1073/pnas.1003428107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Zhan S., Wang T., Wang M., Li J., Ge W. In-depth proteomics analysis to identify biomarkers of papillary thyroid cancer patients older than 45 years with different degrees of lymph node metastases. Proteomics Clin. Appl. 2019;13:e1900030. doi: 10.1002/prca.201900030. [DOI] [PubMed] [Google Scholar]
- 146.Henderson I.C., Allegra J.C., Woodcock T., Wolff S., Bryan S., Cartwright K., Dukart G., Henry D. Randomized clinical trial comparing mitoxantrone with doxorubicin in previously treated patients with metastatic breast cancer. J. Clin. Oncol. 1989;7:560–571. doi: 10.1200/JCO.1989.7.5.560. [DOI] [PubMed] [Google Scholar]
- 147.Allison K.E., Coomber B.L., Bridle B.W. Metabolic reprogramming in the tumour microenvironment: A hallmark shared by cancer cells and T lymphocytes. Immunology. 2017;152:175–184. doi: 10.1111/imm.12777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Li F., Simon M.C. Cancer cells don’t live alone: Metabolic communication within tumor microenvironments. Dev. Cell. 2020;54:183–195. doi: 10.1016/j.devcel.2020.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Lyssiotis C.A., Kimmelman A.C. Metabolic interactions in the tumor microenvironment. Trends Cell Biol. 2017;27:863–875. doi: 10.1016/j.tcb.2017.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Liang J., Shang Y. Estrogen and cancer. Annu. Rev. Physiol. 2013;75:225–240. doi: 10.1146/annurev-physiol-030212-183708. [DOI] [PubMed] [Google Scholar]
- 151.Jameera B.A., Jubie S., Nanjan M.J. Estrogen receptor agonists/antagonists in breast cancer therapy: A critical review. Bioorg. Chem. 2017;71:257–274. doi: 10.1016/j.bioorg.2017.02.011. [DOI] [PubMed] [Google Scholar]





