RT Journal Article SR Electronic T1 Orthosteric-allosteric dual inhibitors of PfHT1 as selective anti-malarial agents JF bioRxiv FD Cold Spring Harbor Laboratory SP 2020.08.25.260232 DO 10.1101/2020.08.25.260232 A1 Jian Huang A1 Yafei Yuan A1 Na Zhao A1 Debing Pu A1 Qingxuan Tang A1 Shuo Zhang A1 Shuchen Luo A1 Xikang Yang A1 Nan Wang A1 Yu Xiao A1 Tuan Zhang A1 Zhuoyi Liu A1 Tomoyo Sakata-Kato A1 Xin Jiang A1 Nobutaka Kato A1 Nieng Yan A1 Hang Yin YR 2020 UL http://biorxiv.org/content/early/2020/08/26/2020.08.25.260232.abstract AB Artemisinin-resistant malaria parasites have emerged and been spreading, posing a significant public health challenge. Anti-malarial drugs with novel mechanisms of action are therefore urgently needed. In this report, we exploit a “selective starvation” strategy by selectively inhibiting Plasmodium falciparum hexose transporter 1 (PfHT1), the sole hexose transporter in Plasmodium falciparum, over human glucose transporter 1 (hGLUT1), providing an alternative approach to fight against multidrug-resistant malaria parasites. Comparison of the crystal structures of human GLUT3 and PfHT1 bound to C3361, a PfHT1-specific moderate inhibitor, revealed an inhibitor binding-induced pocket that presented a promising druggable site. We thereby designed small-molecules to simultaneously block the orthosteric and allosteric pockets of PfHT1. Through extensive structure-activity relationship (SAR) studies, the TH-PF series was identified to selectively inhibit PfHT1 over GLUT1 and potent against multiple strains of the blood-stage P. falciparum. Our findings shed light on the next-generation chemotherapeutics with a paradigm-shifting structure-based design strategy to simultaneously targeting the orthosteric and allosteric sites of a transporter.Significance statement Blocking sugar uptake in P. falciparum by selectively inhibiting the hexose transporter PfHT1 kills the blood-stage parasites without affecting the host cells, indicating PfHT1 as a promising therapeutic target. Here, we report the development of novel small-molecule inhibitors that are selectively potent to the malaria parasites over human cell lines by simultaneously targeting the orthosteric and the allosteric binding sites of PfHT1. Our findings established the basis for the rational design of next-generation anti-malarial drugs.Competing Interest StatementDeclaration of Interests: A patent application was filed. Applicant: Institution: Tsinghua University Application number: PCT/CN2020/074258 Status of application: Not yet published. Specific aspects of manuscript covered in the patent application: Crystal structure of PfHT1 in complex with C3361, the inhibitor binding-induced pocket in C3361-bound structure, the inhibitory activities of TH-PF01 and its derivatives.