首页 | 本学科首页   官方微博 | 高级检索  
     


Catastrophic ATP loss underlies a metabolic combination therapy tailored for MYCN-amplified neuroblastoma
Authors:Krista M. Dalton  Timothy L. Lochmann  Konstantinos V. Floros  Marissa L. Calbert  Richard Kurupi  Giovanna T. Stein  Joseph McClanaghan  Ellen Murchie  Regina K. Egan  Patricia Greninger  Mikhail Dozmorov  Sivapriya Ramamoorthy  Madhavi Puchalapalli  Bin Hu  Lisa Shock  Jennifer Koblinski  John Glod  Sosipatros A. Boikos  Cyril H. Benes  Anthony C. Faber
Abstract:MYCN-amplified neuroblastoma is a lethal subset of pediatric cancer. MYCN drives numerous effects in the cell, including metabolic changes that are critical for oncogenesis. The understanding that both compensatory pathways and intrinsic redundancy in cell systems exists implies that the use of combination therapies for effective and durable responses is necessary. Additionally, the most effective targeted therapies exploit an “Achilles’ heel” and are tailored to the genetics of the cancer under study. We performed an unbiased screen on select metabolic targeted therapy combinations and correlated sensitivity with over 20 subsets of cancer. We found that MYCN-amplified neuroblastoma is hypersensitive to the combination of an inhibitor of the lactate transporter MCT1, AZD3965, and complex I of the mitochondrion, phenformin. Our data demonstrate that MCT4 is highly correlated with resistance to the combination in the screen and lowly expressed in MYCN-amplified neuroblastoma. Low MCT4 combines with high expression of the MCT2 and MCT1 chaperone CD147 in MYCN-amplified neuroblastoma, altogether conferring sensitivity to the AZD3965 and phenformin combination. The result is simultaneous disruption of glycolysis and oxidative phosphorylation, resulting in dramatic disruption of adenosine triphosphate (ATP) production, endoplasmic reticulum stress, and cell death. In mouse models of MYCN-amplified neuroblastoma, the combination was tolerable at concentrations where it shrank tumors and did not increase white-blood-cell toxicity compared to single drugs. Therefore, we demonstrate that a metabolic combination screen can identify vulnerabilities in subsets of cancer and put forth a metabolic combination therapy tailored for MYCN-amplified neuroblastoma that demonstrates efficacy and tolerability in vivo.

Despite their relative rarity compared to blood cancers, solid-tumor pediatric cancers are now the leading cause of pediatric cancer-related deaths. Among the most deadly is high-risk neuroblastoma (NB): amplification of MYCN confers high risk and is the clear driver of NB in these cancers (1). As such, MYCN remains the most important drug target in NB and one of the most important in pediatric cancer. Unfortunately, direct chemical targeting of MYCN has not yet been successful, and despite advancements in anti-GD2 immunotherapy (2), alternate ways of targeting MYCN-amplified NB may be needed to successfully treat this cancer.One approach is to find tumor-specific vulnerabilities, which are exploitable pharmacologically. Many efforts, including ours (3), have exhaustively looked for kinase inhibitors with particular efficacy in MYCN-amplified NBs. However, the emerging picture is a lack of kinase inhibitor efficacy in MYCN-amplified NB. Other vulnerabilities may be classified under the broad category of drugs targeting epigenetic modifiers. For example, using a CRISPR/Cas9 screen, Stegmaier and colleagues demonstrated that MYCN-amplified NB may be susceptible to targeting the H3K27me methylase EZH2 (4); in a different study, they demonstrated the susceptibility of MYCN-amplified NB to the combination of BRD4 inhibitors with CDK7 inhibitors (5). In addition, Thiele and colleagues (6) demonstrated high-risk NBs were susceptible to inhibition of the lysine methyltransferase SETD8. As promising as these data are, it remains unknown whether tolerability and/or clinical activity in MYCN-amplified NB will occur and SETD8, BRD4, and CDK7 inhibitors so far are not in the pediatric clinic. Cell death inducers constitute a third category. To this point, we recently uncovered a susceptibility of MYCN-amplified NB to the BCL-2 inhibitor venetoclax (3), confirmed by others (7). There, MYCN-driven NOXA expression sensitizes cells to venetoclax (3). Venetoclax is now in early phase trials in pediatric patients including those with NB (NCT03236857). It remains to be seen whether or not it will elicit responses in NB patients as a single agent.A fourth distinct category of therapeutic strategies to indirectly target oncogenes is through metabolism targeting, involving the growing coterie of drugs targeting the pathways fulfilling the high-energy demands of cancer cells. A major energy currency in cells is adenosine triphosphate (ATP). The Warburg effect describes the propensity of cancer cells (and highly proliferating normal cells) to produce ATP in the presence of oxygen with the less efficient, extramitochondrial glycolysis, as opposed to the more efficient mitochondria-based oxidative phosphorylation occurring in most noncancerous cells (8). The mechanistic explanation of the Warburg effect and how it might benefit cancer cells has been revised dramatically over the years. It was originally proposed that mitochondria from cancer cells were defective and lacked oxidative phosphorylation capabilities (9); on the contrary, emerging data show that many cancers rely on oxidative phosphorylation to facilitate the generation of ATP (8, 10). Interestingly, while amplified MYCN directly regulates the expression of many of the key glycolytic enzymes and as such contributes to the Warburg effect (11, 12), a study utilizing a Seahorse respirator demonstrated that a MYCN-amplified NB cell line favored oxidative phosphorylation over glycolysis for the metabolic needs, while the reverse was true for a MYCN wild-type NB cell line (13). In an independent study, MYCN was associated with higher glycolytic flux and oxidative phosphorylation and conferred sensitivity to fatty acid oxidation disruption (12). Overall, since c-MYC, which shares ∼40% binding homology to DNA-binding sites throughout the genome with MYCN, has been extensively characterized as a metabolic master regulator (14, 15), it is likely there are other MYCN-driven metabolic processes that may represent significant drug targets.Monocarboxylate transporters (MCTs) consist of four members (MCT1–4) in mammalian cells. Among their most critical substrates are lactate and pyruvate; MCT1 and MCT4 are responsible for lactate export across the plasma membrane to the extracellular space (16). AZD3965 (17) (AstraZeneca) is the first in-class–specific MCT1/2 dual inhibitor and is currently in early phase trials for diverse cancers; however, other inhibitors from different companies have recently been developed as well (18). Of note, AZD3965 has demonstrated good tolerability in diverse patients (clinical trial number NCT01791595). Although rare (65 cases/100,000 person-years), lactic acidosis led to the market retrieval of phenformin in America (19), yet phenformin remains in use as a type II antidiabetic drug in Europe, functioning centrally as a mitochondrial complex I electron transport chain (ETC) inhibitor. Phenformin reduces both glycolytic intermediates and pyruvate, increases shunting of glucose-derived carbon (increasing total lactate production), and markedly reduces tricarboxylic acid cycle intermediates (20). Indeed, there has been a recent resurgence in interest in the use of phenformin to treat cancer. For example, in BRAF mutant melanoma, phenformin sensitized cells to BRAF inhibitor through cooperative suppression of the metabolic sensor pathway mTORC1 (21). These preclinical data have led to a clinical trial of phenformin in combination with BRAF inhibitor in BRAF mutant melanoma (NCT03026517). Overall, while targeting individual metabolic pathways has demonstrated some preclinical success in different cancer models, it is limited with significant redundancy in pathways to generate ATP and regenerate NAD+ (22). We therefore assessed potential combination therapies involving metabolic targeting drugs to identify a strategy for MYCN-amplified NB.
Keywords:neuroblastoma   MYCN   metabolomics   apoptosis   lactate
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号