+ All Categories
Home > Documents > Abstract Number 3588 Identification of a potent, orally ......We have identified MCT4 specific...

Abstract Number 3588 Identification of a potent, orally ......We have identified MCT4 specific...

Date post: 23-Sep-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
1
Identification of a potent, orally bioavailable and selective MCT4 inhibitor for the treatment of solid Warburg tumors N. Hambruch , B. Herkert, C. Gege, A. Mallinger, J. Fabian, Y. Wang, M. Albers, O. Kinzel, G. Fink, F. Braun, A. Funk, A. Bittmann-Waetzig, T. Fleckenstein, M. Birkel & C. Kremoser Results Waldhofer Str. 104, 69123 Heidelberg, Germany Abstract Number 3588 Phenex MCT4 inhibitor screening path 1. Hanahan & Weinberg; Cell (2011) 2. Marchiq & Pouyssegur; J Mol Med Berl. (2015) 3. San-Millan & Brooks; Carcinogenesis (2016) 4. Romero-Garcia et al.; Front. Immunol. (2016) 5. Dhup et al.; Curr. Pharm. Des. (2012) 6. Baumann et al.; Neuro-Oncol. (2009) 7. Brizel et al.; Oncol. Biol. Phys. (2001) 8. Morrot et al.; Front. Oncol. (2018) 9. Buck et al.; Cell (2017) 10.Ullah et al.; J Biol Chem. (2006) References Highly glycolytic solid tumor Introduction We have identified MCT4 specific inhibitors (MCT4i) and significantly improved activity and selectivity of the first hits via an extensive medicinal chemistry program. These efforts resulted in the identification of highly potent, orally bioavailable and selective MCT4i. (MCT4i IC 50 ≤ 20nM with MCT1i IC 50 ≥ 100µM) Activity of MCT4i was significantly improved through chemical modification of initial screening hits. Intracellular lactate accumulation (Lactate-Glo™ assay, Promega) after 6h of inhibitor treatment was analyzed in MDA-MB-231 (MCT4 + ) cells. MCT1i AZD 3965 served as negative control. Selectivity of MCT4 over MCT1/2 was investigated by incubating BT-20 (MCT1 + ) or MCF-7 (MCT1 + /2 + ) cells with the test cpds (data not shown). MCT4i impair growth of highly glycolytic MDA- MB-231 (MCT4 + ) cells. The anti-proliferative effect (CyQuant assay) over 96 h directly correlates with MCT4i activity determined by intracellular lactate accumulation. MCT1i AZD 3965 does not affect the growth of MCT1 - MDA-MB-231 cells. SKBR3 cell lysates were incubated with different concentrations of MCTi for 60min. Afterwards, samples were incubated @ 68°C for 6 min and subjected to western blot analysis of MCT4 or MCT1 protein levels. MCT4i lead to a concentration-dependent stabilization of exclusively MCT4 protein (filled symbol), while MCT1i showed the same effect only on MCT1 protein (open symbol). Vinculin was used as loading control. Mouse PK data of PX-788 shows acceptable profile for pre-clinical mouse studies. MCT4i have no effect on proliferation (A) or lactate accumulation (B) in activated and proliferating CD3 + /CD8 + T cells. Human T cells were isolated from buffy coat using CD4 & CD8 beads. During a 4 day activation period (CD3/CD28/IL2) cells were treated continuously with different MCT4i or MCT1i. Inhibition of MCT1 but not MCT4 leads to reduced proliferation (CFSE intensity determined by FACS) and an intracellular accumulation of lactate (Lactate-Glo™ assay). Tumor growth in NCI-H358 xenograft is impaired by inhibition of MCT4. NOD.SCID mice were inoculated with NCI-H358 cells and treated for 22 days with MCT4i or Cediranib after tumor establishment. Mice displayed no adverse signs and maintained normal body weight over the course of the study. This study was completed shortly before the meeting. The ex vivo analysis of tumor samples is currently ongoing. Cediranib was used as a responsiveness control for the model. Statistics: ANOVA test (A) (B) MCT4 inhibition Establishment of tumor lactate level as PD marker MCT4 is a direct target of Phenex MCT4i MCT4i show no effect on T cell proliferation or lactate accumulation PX-788 mouse PK data MCT4i impair growth of highly glycolytic cells Improvement of Phenex MCT4i activity MCT4i shows single-arm efficacy in NCI-H358 murine xenograft Mann-Whitney test ****p<0.0001 Conclusion & Outlook A phenotypic screening campaign identified several compounds that were inhibitors of lactate efflux via MCT4. Medicinal chemistry efforts led to a significant improvement of MCT4 inhibitor potency and metabolic stability. Cellular target engagement of early tool compounds was confirmed. Optimization of one series yielded PX-788 which shows single-arm efficacy in the NCI-H358 murine xenograft model. Synergy with anti-angiogenic therapies will be evaluated in subsequent studies. Human CD3 + /CD8 + T cell proliferation is not impaired by MCT4 inhibition. Therefore, MCT4 inhibitors represent promising rational combination partners for immuno- modulatory drugs due to the relief of lactate-mediated immunosuppression in the TME. In the future, we will further analyze if MCT4 inhibitors are able to enhance response to checkpoint inhibition in pre- clinical models. Glycolysis Pyruvate OXPHOS Lactate Lactate H + H + Glucose GLUT MCT4 ATP HIF-Hypoxic tumor cell Glycolysis Pyruvate OXPHOS Lactate Lactate H + H + Glucose GLUT MCT4 ATP HIF-Hypoxic tumor cell MCT4i One of the most cited publications in the cancer research field by Hanahan & Weinberg acknowledges metabolic reprogramming as one hallmark of cancer 1 . A key metabolic alteration in tumor cells is an elevated glycolysis rate. While non-transformed cells mainly utilize the citric acid cycle and oxidative phosphorylation for energy production and anabolic growth, tumor cells employ aerobic glycolysis. These alterations of cellular metabolism are by far more common in solid tumors compared to hematological malignancies 2,3 . Due to the enhanced glycolysis rate, cancer cells produce increased amounts of protons and lactate (up to 40-fold) leading to lactic acidosis within the tumor microenvironment (TME) 2,4 . Recent studies support the critical role of this metabolite in tumorigenesis and demonstrate versatile tumor-promoting effects of lactate. As one major contribution the stimulation of angiogenesis is discussed, an effect at least in part mediated through the activation of the VEGF/VEGFR signaling pathway in endothelial cells, which is the central regulator of angiogenesis 5 . Furthermore, lactate has been implicated in cancer cell migration, and intratumoral lactate levels are highly correlated with metastasis in different types of cancers 5-7 . Moreover, lactic acidosis within the tumor microenvironment has been shown to have differential effects on distinct immune cell subpopulations, eventually contributing to tumor immune escape and cancer progression 8,9 . Lactate is transported across the plasma membrane by the monocarboxylate transporters 1 and 4 (MCT1/4) in symport with a proton. While MCT1 is expressed almost ubiquitously at low levels, MCT4 expression is regulated by hypoxia-inducible factor 1α (Hif1α), the central player in the cellular response to hypoxia 10 . Thus, MCT4 is present in hypoxic, highly glycolytic cells. Given their central role in prevention of intracellular acidification and lactate transport, MCTs appear to be indispensable for glycolytic tumor growth. Since MCT4 has the strongest implication on the development and aggressiveness of diverse cancer types it has emerged as a new target for the treatment of solid tumors. Blocking lactate export by inhibition of MCT4 is expected to impair tumor cell metabolism and reduce lactic acidosis in the TME. Recent publications support the notion that this will result in: Tumor cell growth arrest Radio-sensitivity Chemo-sensitivity Restoration of cancer immunosurveillance
Transcript
Page 1: Abstract Number 3588 Identification of a potent, orally ......We have identified MCT4 specific inhibitors (MCT4i) and significantly improved activity and selectivity of the first hits

Identification of a potent, orally bioavailable and selective MCT4 inhibitor for the treatment of solid Warburg tumors N. Hambruch, B. Herkert, C. Gege, A. Mallinger, J. Fabian, Y. Wang, M. Albers, O. Kinzel,

G. Fink, F. Braun, A. Funk, A. Bittmann-Waetzig, T. Fleckenstein, M. Birkel & C. Kremoser

Results

Waldhofer Str. 104, 69123 Heidelberg, Germany

Abstract Number 3588

Phenex MCT4 inhibitor screening path

1. Hanahan & Weinberg; Cell (2011)2. Marchiq & Pouyssegur; J Mol Med Berl. (2015)3. San-Millan & Brooks; Carcinogenesis (2016)4. Romero-Garcia et al.; Front. Immunol. (2016)5. Dhup et al.; Curr. Pharm. Des. (2012)6. Baumann et al.; Neuro-Oncol. (2009)7. Brizel et al.; Oncol. Biol. Phys. (2001)8. Morrot et al.; Front. Oncol. (2018)9. Buck et al.; Cell (2017)10.Ullah et al.; J Biol Chem. (2006)

References

Highly glycolytic solid tumor

Introduction

We have identified MCT4 specific inhibitors (MCT4i) and significantlyimproved activity and selectivity of the first hits via an extensivemedicinal chemistry program. These efforts resulted in the identificationof highly potent, orally bioavailable and selective MCT4i.(MCT4i IC50 ≤ 20nM with MCT1i IC50 ≥ 100µM)

Activity of MCT4i was significantly improved through chemical modification of initial screening hits.Intracellular lactate accumulation (Lactate-Glo™ assay, Promega) after 6h of inhibitor treatment wasanalyzed in MDA-MB-231 (MCT4+) cells. MCT1i AZD 3965 served as negative control. Selectivity ofMCT4 over MCT1/2 was investigated by incubating BT-20 (MCT1+) or MCF-7 (MCT1+/2+) cells withthe test cpds (data not shown).

MCT4i impair growth of highly glycolytic MDA-MB-231 (MCT4+) cells. The anti-proliferativeeffect (CyQuant assay) over 96 h directlycorrelates with MCT4i activity determined byintracellular lactate accumulation. MCT1i AZD3965 does not affect the growth of MCT1-

MDA-MB-231 cells.

SKBR3 cell lysates were incubated withdifferent concentrations of MCTi for 60min.Afterwards, samples were incubated @ 68°Cfor 6 min and subjected to western blotanalysis of MCT4 or MCT1 protein levels.MCT4i lead to a concentration-dependentstabilization of exclusively MCT4 protein (filledsymbol), while MCT1i showed the same effectonly on MCT1 protein (open symbol). Vinculinwas used as loading control.

Mouse PK data of PX-788 shows acceptable profile for pre-clinical mouse studies.

MCT4i have no effect on proliferation (A) or lactate accumulation (B) in activated and proliferating CD3+/CD8+

T cells. Human T cells were isolated from buffy coat using CD4 & CD8 beads. During a 4 day activationperiod (CD3/CD28/IL2) cells were treated continuously with different MCT4i or MCT1i. Inhibition of MCT1 butnot MCT4 leads to reduced proliferation (CFSE intensity determined by FACS) and an intracellularaccumulation of lactate (Lactate-Glo™ assay).

Tumor growth in NCI-H358 xenograft is impaired by inhibition of MCT4. NOD.SCID mice were inoculatedwith NCI-H358 cells and treated for 22 days with MCT4i or Cediranib after tumor establishment. Micedisplayed no adverse signs and maintained normal body weight over the course of the study. This study wascompleted shortly before the meeting. The ex vivo analysis of tumor samples is currently ongoing. Cediranibwas used as a responsiveness control for the model. Statistics: ANOVA test

(A) (B)

MCT4 inhibition

Establishment of tumor lactate level as PD marker

MCT4 is a direct target of Phenex MCT4i

MCT4i show no effect on T cell proliferation or lactate accumulation

PX-788 mouse PK data

MCT4i impair growth of highly glycolytic cells

Improvement of Phenex MCT4i activity

MCT4i shows single-arm efficacy in NCI-H358 murine xenograft

Mann-Whitney test ****p<0.0001

Conclusion & Outlook A phenotypic screening campaign identified several

compounds that were inhibitors of lactate efflux via MCT4.

Medicinal chemistry efforts led to a significant improvementof MCT4 inhibitor potency and metabolic stability.

Cellular target engagement of early tool compounds wasconfirmed.

Optimization of one series yielded PX-788 which showssingle-arm efficacy in the NCI-H358 murine xenograftmodel. Synergy with anti-angiogenic therapies will beevaluated in subsequent studies.

Human CD3+/CD8+ T cell proliferation is not impaired byMCT4 inhibition. Therefore, MCT4 inhibitors representpromising rational combination partners for immuno-modulatory drugs due to the relief of lactate-mediatedimmunosuppression in the TME.

In the future, we will further analyze if MCT4 inhibitors areable to enhance response to checkpoint inhibition in pre-clinical models.

Glycolysis

Pyruvate

OXPHOS

Lactate

Lactate

H+

H+

Glucose

GLUT

MCT4

ATP

HIF-1α

Hypoxic tumor cell

Glycolysis

Pyruvate

OXPHOS

Lactate

Lactate

H+

H+

Glucose

GLUT

MCT4

ATP

HIF-1α

Hypoxic tumor cell

MCT4i

One of the most cited publications in the cancer research field by Hanahan &Weinberg acknowledges metabolic reprogramming as one hallmark ofcancer1. A key metabolic alteration in tumor cells is an elevated glycolysisrate. While non-transformed cells mainly utilize the citric acid cycle andoxidative phosphorylation for energy production and anabolic growth, tumorcells employ aerobic glycolysis. These alterations of cellular metabolism areby far more common in solid tumors compared to hematologicalmalignancies2,3.Due to the enhanced glycolysis rate, cancer cells produce increased amountsof protons and lactate (up to 40-fold) leading to lactic acidosis within the tumormicroenvironment (TME)2,4. Recent studies support the critical role of thismetabolite in tumorigenesis and demonstrate versatile tumor-promotingeffects of lactate. As one major contribution the stimulation of angiogenesis isdiscussed, an effect at least in part mediated through the activation of theVEGF/VEGFR signaling pathway in endothelial cells, which is the centralregulator of angiogenesis5. Furthermore, lactate has been implicated incancer cell migration, and intratumoral lactate levels are highly correlated withmetastasis in different types of cancers5-7. Moreover, lactic acidosis within thetumor microenvironment has been shown to have differential effects ondistinct immune cell subpopulations, eventually contributing to tumor immuneescape and cancer progression8,9.

Lactate is transported across theplasma membrane by themonocarboxylate transporters 1and 4 (MCT1/4) in symport with aproton. While MCT1 is expressedalmost ubiquitously at low levels,MCT4 expression is regulated byhypoxia-inducible factor 1α (Hif1α),the central player in the cellularresponse to hypoxia10. Thus, MCT4is present in hypoxic, highlyglycolytic cells. Given their centralrole in prevention of intracellularacidification and lactate transport,MCTs appear to be indispensablefor glycolytic tumor growth.

Since MCT4 has the strongestimplication on the development andaggressiveness of diverse cancertypes it has emerged as a newtarget for the treatment of solidtumors.Blocking lactate export by inhibitionof MCT4 is expected to impairtumor cell metabolism and reducelactic acidosis in the TME. Recentpublications support the notion thatthis will result in:

Tumor cell growth arrestRadio-sensitivityChemo-sensitivityRestoration of cancer immunosurveillance

Recommended