Skip to main content
Acta Pharmaceutica Sinica. B logoLink to Acta Pharmaceutica Sinica. B
. 2023 Dec 16;14(3):905–952. doi: 10.1016/j.apsb.2023.12.010

Small-molecule agents for cancer immunotherapy

Fang Wang a,, Kai Fu a,, Yujue Wang b, Can Pan a, Xueping Wang a, Zeyu Liu a, Chuan Yang a, Ying Zheng c, Xiaopeng Li d, Yu Lu e, Kenneth Kin Wah To f,, Chenglai Xia g,, Jianye Zhang e,, Zhi Shi d,, Zeping Hu b,, Min Huang c,, Liwu Fu a,
PMCID: PMC10935485  PMID: 38486980

Abstract

Cancer immunotherapy, exemplified by the remarkable clinical benefits of the immune checkpoint blockade and chimeric antigen receptor T-cell therapy, is revolutionizing cancer therapy. They induce long-term tumor regression and overall survival benefit in many types of cancer. With the advances in our knowledge about the tumor immune microenvironment, remarkable progress has been made in the development of small-molecule drugs for immunotherapy. Small molecules targeting PRR-associated pathways, immune checkpoints, oncogenic signaling, metabolic pathways, cytokine/chemokine signaling, and immune-related kinases have been extensively investigated. Monotherapy of small-molecule immunotherapeutic drugs and their combinations with other antitumor modalities are under active clinical investigations to overcome immune tolerance and circumvent immune checkpoint inhibitor resistance. Here, we review the latest development of small-molecule agents for cancer immunotherapy by targeting defined pathways and highlighting their progress in recent clinical investigations.

KEY WORDS: Cancer immunotherapy, Small-molecule agents, Immune checkpoints, Oncogenic signaling, Metabolic pathways, Cytokine/chemokine signaling, Antitumor immunity, Tumor immune microenvironment

Graphical abstract

This review systematically summarizes small-molecule agents for cancer immunotherapy implicated in PRR-associated pathways, immune checkpoint, oncogenic signaling, metabolic pathways, cytokine/chemokine signaling, immune-related kinases, etc.

Image 1

1. Introduction

Cancer immunotherapy, which aims to control and eradicate tumors by reestablishing normal antitumor immune responses, has emerged as a strategy for cancer treatment with enormous potential. The history of cancer immunotherapy could be traced back to 1891 upon an important observation made by Dr. William B. Coley, widely accepted as the “Father of Immunotherapy”. He found that certain bacterial infections in cancer patients could lead to tumor regression or even complete remission by causing necrosis1,2. This astonishing discovery has attracted scientists worldwide to investigate novel strategies to combat cancer. In 1986, the US Food and Drug Administration (FDA) approved an antitumor cytokine called interferon-α (IFNα) as the first immunotherapeutic agent for hairy cell leukemia3. After decades of exploration, the development of ipilimumab (a CTLA-4 monoclonal antibody) and chimeric antigen receptor (CAR) T cell therapy marked a significant breakthrough in the field of cancer immunotherapy in 2013, indicating that cancer treatment has entered the era of immunotherapy4. The advent of cancer immunotherapy has revolutionized cancer treatment, substantially prolonged the survival of cancer patients, and improved their quality of life.

Cancer immunotherapy is comprised of cancer vaccines, adoptive cellular immunotherapy (e.g., CAR-T cell therapy), immunomodulators (e.g., cytokines), and immune checkpoint inhibitors (ICIs), with ICIs being the most widely used and promising drugs. ICIs such as anti-PD-1/PD-L1 antibodies and anti-CTLA4 antibodies show clinical benefits in a range of cancer types, including non-small cell lung cancer (NSCLC), melanoma, renal carcinoma, and breast cancer5, 6, 7. However, only 20%–30% of patients respond to ICIs and the majority of patients fail to benefit from these agents. The low response rates of ICIs are often due to poor tissue permeability and single-target inhibition. In addition, the long half-life and inherent immunogenicity of ICIs could contribute to high rates of immune-related adverse events. ICIs also have several other drawbacks such as high costs and intravenous or subcutaneous route of administration8,9. These clinical challenges hamper the widespread application of ICIs and it is urgent to develop other strategies to improve the clinical efficacy of cancer immunotherapy. To this end, small-molecule agents targeting tumor immunity have been developed for their oral bioavailability, short half-life, membrane permeability, extensive tissue penetration, low immunogenicity, and manageable adverse events, which lead to more potent antitumor activity and safer clinical application8. Improved understanding of tumor immunology has led to the development of small-molecule immunotherapeutics, which could activate or reactivate the immune system to attack cancer cells. Numerous small-molecule agents implicated in pattern recognition receptor-associated pathways, immune checkpoints, oncogenic signaling, metabolic pathways, cytokine/chemokine signaling, and immune-related kinases have been studied in the past few years. A substantial number of small-molecule drugs have entered clinical trials and some of them have received clinical approval for cancer treatment.

This review summarizes the recent advancements in the development of small molecule-based cancer immunotherapy, with special emphasis on those under clinical investigation and on the market (Table 1, Table 2).

Table 1.

Small-molecule agents under clinical development in immuno-oncology.

Target pathway Agent Company Function Stage NCT number
cGAS/STING ADU-S100/MIW815 Chinook Therapeutics CDN agonist Phase I NCT02675439
BMS-986301 Bristol-Myers Squibb CDN agonist Phase I NCT03956680
SB-11285 F-star Therapeutics CDN agonist Phase I NCT04096638
TAK-676 Takeda CDN agonist Phase I NCT04420884
BI 1387446 Boehringer Ingelheim CDN agonist Phase I NCT04147234
GSK3745417 GlaxoSmithKline Non-CDN agonist Phase I NCT03843359
E7766 Eisai Macrocycle-bridged CDN Phase I NCT04144140
SNX281 Stingthera Non-CDN agonist Phase I NCT04609579
MK-2118 Merck Sharp & Dohme Non-CDN agonist Phase I NCT03249792
IMSA101 ImmuneSensor Therapeutics cGAMP analogue Phase I/IIa NCT04020185
KL340399 Sichuan Kelun Non-CDN agonist Phase I NCT05387928, NCT05549804
HG381 HitGen Non-CDN agonist Phase I NCT04998422
CDK-002 (exoSTING) Codiak BioSciences Exsome agonist Phase I/II NCT04592484
TLRs Amplivant Leiden University TLR2 agonist Phase I NCT02821494
Poly-ICLC (Hiltonol) Oncovir TLR3 agonist Phase II NCT02423863
IDC-G305 Immune Design TLR4 agonist Phase I NCT02015416
Entolimod Roswell Park Cancer Institute TLR5 agonist Phase I NCT01527136
Mobilan (M-VM3) Panacela Labs TLR5 agonist Phase I NCT02654938
Imiquimod Medical University of Graz, United States Naval Medical Center, VA Office of Research and Development, Barretos Cancer Hospital, NYU Langone Health Medical University of Vienna, Medical University of South Carolina TLR7 agonist Phase III NCT01861535
Phase II/III NCT02130323
Phase III NCT02059499
Phase III NCT05212246
Phase II NCT03233412
Phase II NCT00899574
Phase II NCT00941811
Phase I NCT04883645
TMX-101 Telormedix SA TLR7 agonist Phase II NCT01731652
DSP-0509 Sumitomo Pharma Oncology TLR7 agonist Phase I/II NCT03416335
BNT411 BioNTech SE TLR7 agonist Phase I/II NCT04101357
RO7119929 Hoffmann-La Roche TLR7 agonist Phase I NCT04338685
LHC165 Novartis TLR7 agonist Phase I NCT03301896
Motolimod (VTX-2337) Celgene TLR8 agonist Phase I NCT03906526
Tilsotolimod (IMO-2125) A.J.M. van den Eertwegh TLR9 agonist Phase II NCT04126876
Lefitolimod (MGN 1703) Mologen TLR9 agonist Phase II NCT02200081
1018 ISS Dynavax TLR9 agonist Phase I NCT00403052
BCG Southwest Oncology Group Multi-TLR agonist Phase III NCT03091660
PD-1/PD-L1 GS-4224 Gilead Sciences PD-L1 antagonist Phase I NCT04049617
INCB086550 Incyte PD-L1 antagonist Phase I NCT03762447
Phase II NCT04629339
ASC61 Gannex Pharma PD-L1 antagonist Phase I NCT05287399
IMMH-010 Tianjin Chasesun Pharmaceutical PD-L1 antagonist Phase I NCT04343859
MAX-10181 Maxinovel Pty PD-L1 antagonist Phase I NCT05196360
Phase I NCT04122339
BPI-371153 Betta Pharmaceuticals PD-L1 antagonist Phase I NCT05341557
VISTA CA170 BMS & Aurigene PD-L1/VISTA antagonist Phase I NCT02812875
TβRI Galunisertib Eli Lilly & Co. TβRI inhibitor Phase I/II NCT02408744
Phase I/II NCT03470350
Phase I NCT02304419
Phase I NCT01722825
Vactosertib Ewha Womans University TβRI inhibitor Phase I NCT03074006
Phase I/II NCT03698825
Phase II NCT04103645
LY3200882 Eli Lilly & Co. TβRI inhibitor Phase I/II NCT04031872
TP-0184 Tolero Pharmaceuticals Inc. TβRI inhibitor Phase I/II NCT04623996
Phase I NCT03429218
GFH018 Genfleet TβRI inhibitor Phase I NCT05051241
CSF1R Pexidartinib Daiichi Sankyo CSF1R, c-KIT, FLT3 Phase III NCT04488822
Surufatinib Hutchison Medipharma CSF-1R, VEGFR1–3, FGFR1 Phase II NCT05171439
Phase II NCT02614495
Phase III NCT02588170
Chiauranib Chipscreen Biosciences CSF1R, VEGFR1–3, PDGFRα, c-Kit Phase I/II NCT03166891
Phase I/II NCT03245190
Phase II NCT05497843
Phase III NCT04830813
Vimseltinib Deciphera Pharmaceuticals CSF1R inhibitor Phase I/II NCT03069469
Phase III NCT05059262
JNJ-40346527 Johnson Pharmaceuticals CSF1R inhibitor Phase I NCT03177460
Phase I NCT01572519
Phase II NCT03557970
ARRY-382 Array Biopharma CSF1R inhibitor Phase I NCT01316822
Chemokine receptor Plozalizumab Millennium Pharmaceuticals CCR2 antagonist Phase I NCT02723006
BMS-813160 Bristol-Myers Squibb CCR2/5 antagonist Phase I/II NCT03496662
Phase I/II NCT03767582
Arginine metabolism INCB001158 Incyte ARG1 antagonist Phase II NCT02903914
Phase II NCT03910530
IDO1 Epacadostat Incyte IDO1 inhibitor Phase III NCT02752074
Phase III NCT03361865
Indoximod NewLink IDO1 inhibitor Phase II NCT01560923
Linrodostat Flexus/BMS IDO1 inhibitor Phase III NCT03329846
Prostaglandin pathway SC-58635 Pfizer COX2 inhibitor Phase III NCT02429427
Phase II NCT00527982
Phase II NCT01158534
Phase II NCT00499655
ONO-4578 Ono Pharmaceutical Co., Ltd. EP4 antagonist Phase I NCT03155061
E7046 Adlai Nortye Biopharma Co., Ltd. EP4 antagonist Phase I NCT02540291
Phase I NCT04432857
Adenosine receptor Arcus Arcus Biosciences CD73 inhibitor Phase II NCT04660812
Phase II NCT04381832
ORIC-533 ORIC Pharmaceuticals CD73 inhibitor Phase I NCT05227144
CS3005 CStone Pharmaceuticals A2aR inhibitor Phase I NCT04233060
EXS21546 Exscientia Limited A2aR inhibitor Phase I NCT04727138
INCB106385 Incyte Corporation A2aR inhibitor Phase I NCT04580485
TT-10 Tarus Therapeutics A2aR inhibitor Phase I/II NCT04969315
Ciforadenant Corvus A2aR inhibitor Phase I NCT03454451
Phase I NCT02655822
Phase I/II NCT05501054
Inupadenant iTeos A2aR inhibitor Phase I NCT05117177
Phase II NCT05403385
Phase I/II NCT05060432
AZD4635 AstraZeneca A2aR inhibitor Phase I/II NCT03381274
Phase I NCT04478513
Taminadenant (PBF-509, NIR178) Novartis Pharmaceuticals A2aR inhibitor Phase I NCT02403193
Phase I NCT03786484
Phase II NCT03207867
Preladenant (MK-3814, SCH-420814) Merck A2aR inhibitor Phase Ib/II NCT03099161
Etrumadenant Arcus A2aR/A2bR inhibitor Phase II NCT05177770
NCT04262856
PI3K Eganelisib Infinity Pharmaceuticals PI3Kγ inhibitor Phase II NCT03795610
Idelalisib Gilead Sciences PI3Kδ inhibitor Phase II NCT01282424
Phase II NCT01393106
Umbralisib TG Therapeutics PI3Kδ/CK1ε inhibitor Phase II NCT02742090
Parsaclisib Incyte PI3Kδ inhibitor Phase II NCT04434937
Phase II NCT02998476
Zandelisib MEI Pharma PI3Kδ inhibitor Phase II NCT03768505
Linperlisib Shanghai YingLi Pharmaceutical PI3Kδ inhibitor Phase II NCT05274997
Copanlisib Bayer PI3Kα/δ inhibitor Phase III NCT02369016
Taselisib Roche Group PI3Kα/δ inhibitor Phase II NCT02785913
Duvelisib Verastem PI3Kγ/δ inhibitor Phase III NCT02004522
Tenalisib Rhizen Pharmaceuticals PI3Kγ/δ inhibitor Phase II NCT03711578
Phase II NCT04204057
Phase II NCT05021900
Buparlisib Adlai Nortye Pan-PI3K inhibitor Phase II NCT01790932
Pictilisib Roche; Genentech Pan-PI3K inhibitor Phase I NCT00876109
JAK Ruxolitinib Incyte JAK1/2 inhibitor Phase II NCT03153982
Phase II NCT03722407
Phase III NCT00952289
Phase III NCT00934544
Fedratinib TargeGen JAK2 inhibitor Phase II NCT05177211
Phase III NCT01437787
Phase II NCT01523171
Pacritinib Cell Therapeutics JAK2 inhibitor Phase I/II NCT04520269
Phase II NCT04635059
Phase II NCT02277093
Phase III NCT01773187
Phase III NCT02055781
Momelotinib Sierra Oncology JAK1/2 inhibitor Phase III NCT01969838
Phase III NCT02101268
Phase III NCT04173494
HPK-1 CFI-402411 Treadwell Therapeutics, Inc. HPK-1 inhibitor Phase I/II NCT04521413
BGB-15025 BeiGene HPK-1 inhibitor Phase I NCT04649385
PF-07265028 Pfizer HPK-1 inhibitor Phase I NCT05233436
PRJ1-3024 Zhuhai Yufan Biotechnologies Co. HPK-1 inhibitor Phase I/II NCT05315167
NDI-101150 Nimbus Saturn, Inc. HPK-1 inhibitor Phase I/II NCT05128487
RON BMS 777607 (ASLAN002) Bristol-Myers Squibb RON/c-Met inhibitor Phase I/II NCT00605618
Aslan Pharmaceuticals RON/c-Met inhibitor Phase I NCT01721148
RORγt LYC-55716 Lycera RORγt agonist Phase II NCT02929862
Bromodomain ABBV-075 AbbVie Bromodomain inhibitor Phase I NCT02391480
Alobresib Gilead Sciences Bromodomain inhibitor Phase I/II NCT02607228
Phase I NCT02392611
BAY 1238097 Bayer Bromodomain inhibitor Phase I NCT02369029
Birabresib Merck Sharp & Dohme LLC Bromodomain inhibitor Phase I NCT02698189
Phase I NCT02698176
Phase II NCT02296476
Phase I NCT02259114
Phase I NCT01713582
BMS-986158 Bristol-Myers Squibb Bromodomain inhibitor Phase I/II NCT02419417
BMS-986378 Bristol-Myers Squibb Bromodomain inhibitor Phase I NCT03936465
CC-90010 Celgene Bromodomain inhibitor Phase I NCT03220347
CCS1477 CellCentric Ltd. Bromodomain inhibitor Phase I/II NCT04068597
Phase I/II NCT03568656
CPI-0610 Constellation Pharmaceuticals Bromodomain inhibitor Phase II NCT02986919
Phase I/II NCT02158858
Phase I NCT02157636
Phase I NCT01949883
GSK2820151 GlaxoSmithKline Bromodomain inhibitor Phase I NCT02630251
GSK525762 GlaxoSmithKline Bromodomain inhibitor Phase II NCT01943851
Phase I NCT01587703
INCB054329 Incyte Corporation Bromodomain inhibitor Phase I/II NCT02431260
INCB057643 Incyte Corporation Bromodomain inhibitor Phase I/II NCT02711137
RO6870810 Hoffmann-La Roche Bromodomain inhibitor Phase I NCT03068351
Phase I NCT02308761
Phase I NCT01987362
SYHA1801 CSPC ZhongQi Pharmaceutical Technology Co., Ltd. Bromodomain inhibitor Phase I NCT04309968
TQB3617 Chia Tai Tianqing Pharmaceutical Group Co., Ltd. Bromodomain inhibitor Phase I NCT05110807
ZEN003694 Zenith Epigenetics Bromodomain inhibitor Phase II NCT05607108
Phase I NCT02705469
DHODH RP7214 Rhizen Pharmaceuticals SA DHODH inhibitor Phase I/II NCT05246384
BAY2402234 Bayer DHODH inhibitor Phase I NCT03404726
AG-636 Agios Pharmaceuticals, Inc. DHODH inhibitor Phase I NCT03834584
ASLAN003 Aslan Pharmaceuticals DHODH inhibitor Phase II NCT03451084
Leflunomide Joseph Sparano, Icahn School of Medicine at Mount Sinai DHODH inhibitor Phase I/II NCT03709446
University Hospital, Basel, Switzerland Not applicable NCT05605587
City of Hope Medical Center, National Cancer Institute (NCI) Phase I/II NCT02509052
PTC299 PTC Therapeutics DHODH inhibitor Phase I NCT03761069

Table 2.

Small-molecule agents in combinational therapies.

Target Agent Combination NCT number Indication Status/phase
cGAS/STING ADU-S100 +Ipilimumab NCT02675439 Advanced/metastatic solid tumors Phase I, Terminated
+PDR001 NCT03172936 Advanced solid tumors or lymphomas Phase I, Terminated
+Pembrolizumab NCT03937141 Head and neck squamous cell carcinoma Phase II, Terminated
MK-1454 +Pembrolizumab NCT03010176 Advanced solid tumors or lymphomas Phase I, Completed
+Pembrolizumab NCT04220866 Head and neck squamous cell carcinoma Phase II, Completed
BMS-986301 +Nivolumab
+Ipilimumab
NCT03956680 Advanced solid tumors Phase I, Active, not recruiting
SB 11285 +Atezolizumab NCT04096638 Solid tumors Phase I, Recruiting
TAK-676 +Radiation
+Pembrolizumab
NCT04879849 Non-small cell lung cancer, Triple negative breast cancer, Head and neck squamous cell carcinoma Phase I, Recruiting
+Pembrolizumab
/Chemotherapy
NCT04420884 Advanced solid tumors Phase I, Recruiting
BI 1387446 +BI 754091 NCT04147234 Advanced solid tumors Phase I, Active, not recruiting
GSK3745417 +Dostarlimab NCT03843359 Advanced solid tumors Phase I, Active, not recruiting
SNX281 +Pembrolizumab NCT04609579 Advanced solid tumors, lymphomas Phase I, Recruiting
MK-2118 +Pembrolizumab NCT03249792 Advanced solid tumors, lymphomas Phase I, Completed
IMSA101 +ICIs NCT04020185 Advanced solid tumors Phase I/II, Recruiting
TLRs XS15 +Multi-peptide vaccine
+Ibrutinib
NCT04688385 Chronic lymphocytic lymphoma Phase I, Recruiting
Poly-ICLC (Hiltonol) +Vaccine
+GM-CSF
+Chemotherapy
NCT02873819 Squamous cell carcinoma of the oral cavity Phase II, Completed
+Vaccine NCT01204684 Brain tumors Phase II, Active, not recruiting
+Vaccine
+IFA
NCT01079741 Melanoma Phase I/II, Completed
+Vaccine
+Bevacizumab
NCT02078648 Glioblastoma Phase I/II, Completed
+Durvalumab
+Tremelimumab
NCT02643303 Solid tumors Phase I/II, Completed
+Vaccine
+Tetanus peptide
+IFA
NCT02126579 Melanoma Phase I/II, Unknown
+Vaccine
+GuaDecitabine
+Atezolizumab
NCT03206047 Recurrent ovarian, Fallopian tube, Primary peritoneal cancer Phase I/II, Active, not recruiting
+rhuFlt3L
/CDX-301
+Radiotherapy
NCT01976585 B-cell lymphoma Phase I/II, Completed
+Vaccine
+Anti-CD40
NCT04364230 Melanoma Phase I/II, Recruiting
+Pembrolizumab NCT02834052 Metastatic Colorectal cancer Phase I/II, Completed
+Vaccine
+Chemotherapy
NCT01834248 Acute myeloid leukemia, Myelodysplastic syndrome Phase I, Completed
+Vaccine
+Nivolumab
+Chemotherapy
NCT03358719 Acute myeloid leukemia Phase I, Completed
+Vaccine
+Pembrolizumab
+Chemotherapy
NCT03380871 Lung cancer Phase I, Completed
+Vaccine
+IFA
NCT01585350 Melanoma Phase I, Completed
+Vaccine NCT01677962 Pancreatic adenocarcinoma Phase I, Completed
+Vaccine
+Lenalidomide
NCT02721043 Solid tumors Phase I, Completed
+Vaccine
+Nivolumab
NCT02897765 Solid tumors Phase I, Completed
+Vaccines
+Tadalafil
NCT02544880 HNSCC Phase I, Completed
+Vaccine
+Citarinostat
+Lenalidomide
NCT02886065 Multiple myeloma Phase I, Completed
+Vaccine
+Durvalumab
NCT02826434 Breast cancer Phase I, Active, not recruiting
+Vaccine
+Pembrolizuma
NCT03362060 Breast cancer Phase I, Active, not recruiting
+Peptide vaccine
+anti-CD27
NCT02924038 Glioma Phase I, Active, not recruiting
+Vaccine
+Nivolumab
NCT02960230 Glioma Phase I/II, Active, not recruiting
+Vaccine
/neoadjuvant
NCT02549833 Glioma Phase I, Active, not recruiting
+Vaccine NCT03300817 Lung cancer Phase I, Active, not recruiting
+Neoadjuvant NCT03262103 Prostate cancer Phase I, Completed
Rintatolimod +Vaccines
+IFA
NCT01312389 Recurrent ovarian, Fallopian tube, Primary peritoneal cancer Phase I/II, Terminated
+Chemokine NCT03403634 Colorectal cancer with liver metastasis Phase II, Completed
+Pembrolizumab
+Chemotherapy
NCT03734692 Recurrent platinum-sensitive ovarian cancer Phase I/II, Recruiting
+IFNα-2b NCT04379518 Cancer patients with mild or moderate COVID-19 infection Phase I/II, Suspended
BO-112 +Pembrolizumab NCT04570332 Melanoma Phase II, Active, not recruiting
GLA-SE (G100) +Vaccine NCT02320305 Melanoma Phase I, Active, not recruiting
+Radiotherapy NCT02035657 Merkel cell carcinoma Phase I, Completed
+Radiotherapy NCT02180698 Sarcoma Phase I, Completed
GSK1795091 +Pembrolizumab
/GSK3174998
/GSK3359609
NCT03447314 Solid tumors Phase I, Completed
TriMix DC +Vaccine NCT01530698 Melanoma Phase I/II, Completed
Imiquimod +Paracetamol
+Iidocaine
NCT02385188 Vulvar Paget disease Phase III, Completed
+Curettage NCT02242929 Basal cell carcinoma Phase III, Active, not recruiting
+Radiation or
+Chemotherapy
NCT01421017 Breast cancer Phase II, Completed
+Sonidegib NCT03534947 Basal cell carcinoma Phase II, Recruiting
+Vaccine NCT02864147 Cervical intraepithelial neoplasia Phase II, Active, not recruiting
+Vaccine
+Lenalidomide
NCT02802943 Chronic lymphocytic lymphoma Phase II, Completed
+Vaccine NCT03180684 VIN2/3 and vulvar HSIL Phase II, Completed
+Vaccine
+Chemotherapy
+Sargramostim
NCT02276300 Gastric cancer, Breast cancer Phase I, Completed
+Chemotherapy/Radiotherapy/Vaccine NCT01678352 Glioma (grade II) Phase I, Completed
+Vaccine NCT02454634 Glioma Phase I, Completed
+Vaccine
+IFA
NCT02293707 Prostate carcinoma Phase II, Completed
+Vaccines
+Chemotherapy
NCT02234921 Prostate carcinoma Phase I, Completed
+Surgery NCT05055050 Bladder cancer Phase I, Recruiting
+Zalifrelimab NCT05375903 Recurrent bladder cancer Phase I, Recruiting
+Chemotherapy NCT03196180 CIN Phase I, Active, not recruiting
+Ultrasound ablation/Pembrolizumab/Atezolizumab NCT04116320 Solid tumors Phase I, Recruiting
+Vaccine
+Chemotherapy
+Nivolumab
+Pembrolizumab
NCT03872947 Solid tumors Phase I, Recruiting
+Chemotherapy NCT03370406 Squamous cell carcinoma Phase I, Recruiting
+Vaccine
+Toripalimab
+GM-CSF
NCT04072900 Melanoma Phase I, Unknown
+Pembrolizumab NCT03276832 Melanoma Phase I, Active, not recruiting
+Vaccine
+hP1A8
NCT04642937 Recurrent glioblastoma Phase I, Active, not recruiting
+Vaccine
+Atezolizumab
NCT04808245 H3-mutated glioma Phase I, Recruiting
SHR2150 +Chemotherapy
+anti-PD-1/anti-CD47
NCT04588324 Metastatic solid tumors Phase I/II, Unknown
DSP-0509 +Pembrolizumab NCT03416335 Tumors Phase I/II, Completed
BNT411 +Chemotherapy
/Atezolizumab
NCT04101357 Solid tumors Phase I/II, Recruiting
RO7119929 +Tocilizumab NCT04338685 Hepatocellular carcinoma,
Biliary tract cancer
Phase I/II, Completed
LHC165 +PDR001 NCT03301896 Solid tumors Phase I/II, Terminated
Resiquimod +Vaccine NCT01204684 Brain tumors Phase II, Active, not recruiting
+Vaccine
+IFA
NCT00821652 Melanoma Phase I/II, Completed
+Vaccine/IFA NCT02126579 Melanoma Phase I/II, Unknown
Motolimod (VTX-2337) +Chemotherapy NCT01666444 Epithelial ovarian cancer,
Fallopian tube cancer,
Primary peritoneal cancer
Phase II, Completed
+Chemotherapy
+Cetuximab
NCT01836029 Squamous cell carcinoma of the head and neck Phase II, Completed
+Durvalumab
+Chemotherapy
NCT02431559 Ovarian cancer Phase I/II, Completed
+Nivolumab NCT03906526 Head and neck squamous cell carcinoma Phase I, Terminated
+Cetuximab NCT01334177 Squamous cell cancer of head and neck Phase I, Completed
+chemotherapy NCT01294293 Epithelial ovarian cancer,
Fallopian tube cancer,
Primary peritoneal cancer
Phase I, Completed
CMP-001 +Nivolumab NCT04695977 Melanoma Phase II/III, Active, not recruiting
+Avelumab
+Utomilumab
+PF-04518600
NCT02554812 Advanced cancer Phase I/II, Terminated
+Nivolumab NCT04401995 Melanoma Phase II, Recruiting
+Pembrolizumab NCT04708418 Melanoma Phase II, Recruiting
+Pembrolizumab NCT04633278 Head and neck squamous cell carcinoma Phase II, Active, not recruiting
+Nivolumab NCT04698187 Melanoma Phase II, Active, not recruiting
+Cemiplimab-rwlc NCT04916002 Multiple tumor types Phase II, Recruiting
+Radiotherapy NCT04807192 Triple-negative breast cancer Phase II, Recruiting
+Nivolumab NCT03618641 Melanoma,
Lymphoma
Phase II, Active, not recruiting
+Ipilimumab
+Anti-OX40
NCT04387071 Advanced pancreatic cancer,
Solid tumors
Phase I/II, Terminated
+Pembrolizumab NCT03983668 Lymphoma Phase I/II, Recruiting
SD-101 +Pembrolizumab
+Radiotherapy
NCT03007732 Prostate cancer Phase II, Active, not recruiting
+Ipilimumab
+Radiotherapy
NCT02254772 Lymphoma Phase I/II, Completed
+Epacadostat
+Radiotherapy
NCT03322384 Solid tumors, lymphoma Phase I/II, Completed
+Ibrutinib
+Radiotherapy
NCT02927964 Lymphoma Phase I/II, Active, not recruiting
+BMS-986178
+Radiotherapy
NCT03410901 Lymphoma Phase I, Active, not recruiting
+Nivolumab
+Radiotherapy
NCT04050085 Pancreatic adenocarcinoma Phase I, Active, not recruiting
+BMS 986178 NCT03831295 Solid tumors Phase I, Active, not recruiting
+Nivolumab/Ipilimumab NCT04935229 Liver metastatic uveal melanoma Phase I, Recruiting
PF-03152676 +Vaccine
+ACT
+Rituximab
+Chemotherapy
+Filgrastim
NCT00490529 Mantle cell lymphoma Phase II, Completed
CpG7910 +Radiotherapy NCT00185965 Recurrent lymphomas Phase I/II, Completed
Tilsotolimod (IMO-2125) +Ipilimumab
+Nivolumab
NCT03865082 Advanced solid tumors Phase II, Active, not recruiting
+Ipilimumab
+Nivolumab
NCT04270864 Advanced solid tumors Phase I, Active, not recruiting
Lefitolimod (MGN1703) +Ipilimumab NCT02668770 Melanoma Phase I, Active, not recruiting
BCG +Lenalidomide NCT01373294 Bladder cancer Phase II, Completed
+Vaccine NCT02015104 Bladder cancer Phase II, Completed
+Sunitinib NCT00794950 Lower urinary tract urothelial carcinoma Phase II, Completed
+Nivolumab/BMS-986205 NCT03519256 Bladder cancer Phase II, Terminated
+ALT803 NCT03022825 Bladder cancer Phase II/III, Recruiting
+Atezolizumab NCT02792192 Bladder cancer Phase I/II, Terminated
+ALT803 NCT02138734 Bladder cancer Phase I/II, Recruiting
+Durvalumab NCT03317158 Bladder cancer Phase I/II, Recruiting
+Rapamycin NCT02753309 Bladder cancer Phase I, Completed
+Pembrolizumab NCT02808143 Bladder cancer Phase I, Active, not recruiting
+Chemotherapy
+RFA
+GM-CSF
NCT04062721 CRC with liver metastasis Phase I, Not yet recruiting
VISTA CA170 +Enzalutamide
+Bicalutamide
NCT01288911 Prostatic neoplasms Phase II, Completed
TβRI Galunisertib +Durvalumab NCT02734160 Pancreatic cancer Phase I, Completed
+Nivolumab NCT02423343 Non-small cell lung cancer, Hepatocellular carcinoma Phase I/II, Completed
+Enzalutamide NCT02452008 Prostate cancer Phase II, Recruiting
+Sorafenib NCT02240433 Hepatocellular carcinoma Phase I, Completed
+Capecitabine NCT05700656 Colorectal cancer Phase I/II, Not yet recruiting
+Lomustine NCT01582269 Glioblastoma Phase II, Active, not recruiting
+Paclitaxel
+Carboplatin
NCT03206177 Ovarian carcinosarcoma Phase I, Active, not recruiting
Vactosertib +Pomalidomide NCT03143985 Multiple myeloma Phase I, Active, not recruiting
+Durvalumab NCT04893252 Stomach neoplasm Phase II, Recruiting
+Paclitaxel NCT03698825 Metastatic gastric cancer Phase I/II, Unknown
+Durvalumab NCT03732274 Non-small cell lung cancer Phase I/II, Unknown
+Pembrolizumab NCT03724851 Metastatic colorectal or gastric cancer Phase I/II, Active, not recruiting
LY3200882 +Pembrolizumab NCT04158700 Advanced cancer Phase I/II, Withdrawn
+Capecitabine NCT04031872 Colorectal cancer Phase I/II, Unknown
GFH018 +Toripalimab NCT04914286 Advanced solid tumor Phase I/II, Recruiting
YL-13027 +Sintilimab NCT05457517 Solid tumors Phase I/II, Recruiting
CSF1R, c-KIT, FLT3 Pexidartinib +Binimetinib NCT03158103 Gastrointestinal stromal tumor Phase I, Completed
+Paclitaxel NCT01525602 Solid tumors Phase I, Completed
+Durvalumab NCT02777710 Solid tumors Phase I, Completed
+Sunitinib NCT02401815 Gastrointestinal stromal tumor Phase I/II, Completed
+Radiation
+Temozolomide
NCT01790503 Glioblastoma Phase I/II, Completed
+Pembrolizumab NCT02452424 Solid tumors Phase I/II, Terminated
+Sirolimus NCT02584647 Sarcoma Phase I/II, Recruiting
CSF-1R, VEGFR1–3, FGFR1 Surufatinib +Tislelizumab NCT05746728 Breast cancer Phase I/II, Not yet recruiting
+Gemcitabine NCT05093322 Solid tumors Phase I/II, Active, not recruiting
+Pamiparib NCT05652283 Ovarian cancer Phase II, Recruiting
+Envafolimab NCT05722977 Sarcoma Phase II, Not yet recruiting
+Vinorelbine NCT04922658 Non-small cell lung cancer Phase II, Recruiting
+Toripalimab NCT05030246 Solid tumors Phase II, Recruiting
+Capecitabine NCT03873532 Biliary tract cancer Phase II/III, Unknown
+Toripalimab NCT05015621 Neuroendocrine carcinoma Phase III, Recruiting
CSF1R, VEGFR1–3, PDGFRα, c-Kit Chiauranib +Capecitabine NCT05336721 Breast cancer Phase II, Recruiting
+Etoposide
+Paclitaxel
NCT03901118 Ovarian cancer Phase II, Completed
CSF1R Vimseltinib +Avelumab NCT04242238 Sarcoma Phase I, Active, not recruiting
+Pembrolizumab NCT02880371 Solid tumors Phase I/II, Terminated
+Spartalizumab NCT02829723 Solid tumors Phase I/II, Terminated
CCR5 Leronlimab +Carboplatin NCT03838367 Triple negative breast neoplasms Phase I/II, Active, not recruiting
Maraviroc +Pembrolizumab NCT03274804 Metastatic colorectal cancer Phase I, Completed
+Chemotherapy NCT03631407 Colorectal neoplasms Phase II, Completed
Vicriviroc +Pembrolizumab NCT03631407 Colorectal neoplasms Phase II, Completed
MK-7690 +Pembrolizumab NCT00976378 Microsatellite stable colorectal cancer Phase I, Completed
CCR2/5 BMS-813160 +Nivolumab NCT04123379 Non-small cell lung cancer, Hepatocellular carcinoma Phase II, Recruiting
CXCL8 BMS-986253 +Nivolumab NCT03689699 Prostate cancer Phase I/II, Active, not recruiting
NCT04050462 Hepatocellular carcinoma Phase II, Active, not recruiting
NCT03400332 Melanoma Phase I/II, recruiting
NCT04572451 Melanoma, Renal cell carcinoma, Unresectable solid tumors Phase I, recruiting
NCT04123379 Non-small cell lung cancer, Hepatocellular carcinoma Phase II, Recruiting
NCT04848116 Head and neck squamous cell carcinoma Phase II, Recruiting
ARG1 INCB001158 +Pembrolizumab NCT02903914 Advanced/metastatic solid tumors Phase I/II, Completed
IDO1 Epacadostat +Pembrolizumab NCT02752074 Metastatic melanoma Phase III, Completed
+Pembrolizumab NCT03361865 Urothelial carcinoma Phase III, Completed
Prostaglandin pathway Celecoxib (SC-58635) +Toripalimab NCT03926338 Colorectal cancer Phase I/II, Recruiting
+5-fluorouracil
/oxaliplatin
/leucovorin
NCT01150045 Colorectal cancer Phase III, Active, not recruiting
+Atorvastatin NCT01220973 Prostate cancer Phase II, Completed
+Erlotinib hydrochloride NCT00499655 Non-small cell lung cancer Phase II, Completed
ONO-4578 +Nivolumab NCT03155061 Advanced or metastatic solid tumors Phase I, Active, not recruiting
E7046 +Pelvic radiotherapy + Capecitabine/folinic acid/5-FU/oxaliplatin (mFOLFOX-6) NCT03152370 Rectal cancer Phase I, Completed
+Atezolizumab NCT04975958 Locally advanced solid tumor Phase I, Recruiting
+Pembrolizumab NCT04432857 Advanced solid tumors Phase I, Recruiting
CR6086 +Balstilimab NCT05205330 Colorectal cancer Phase I/II, Active, not recruiting
A2aR Etrumadenant +Zimberelimab
+Domvanalimab
NCT04791839 Lung cancer Phase II, Recruiting
Ciforadenant +CPI-006 NCT03454451 Non-small cell lung cancer, Renal cell cancer, Colorectal cancer Phase I, Active, not recruiting
+Atezolizumab NCT02655822 Renal cell cancer, Prostate cancer Phase I, Completed
+lpilimumab/nivolumab NCT05501054 Renal cell carcinoma Phase I/II, Recruiting
Inupadenant +Chemotherapy NCT05403385 Non-small cell lung cancer Phase II, Recruiting
taminadenant +PDR001 NCT02403193 Non-small cell lung cancer Phase I, Completed
+DFF332/spartalizumab NCT04895748 Renal cell carcinoma Phase I, Recruiting
AZD4635 +Durvalumab
+Oleclumab
NCT04089553 Prostate cancer Phase II, Active, not recruiting
PI3Kα Alpelisib +Fulvestrant NCT02437318 Breast cancer Phase III, Active, not recruiting
+Trastuzumab
+Pertuzumab
NCT04208178 Breast cancer Phase III, Recruiting
+Olaparib NCT04729387 Ovarian cancer Phase III, Recruiting
+Capecitabine NCT04753203 Colorectal cancer Phase I/II, Not yet recruiting
+Paclitaxel NCT04526470 Gastric cancer Phase I/II, Recruiting
+Tipifarnib NCT04997902 Head and neck squamous cell carcinoma Phase I/II, Recruiting
Inavolisib +Giredestrant NCT05708235 Breast cancer Phase II, Not yet recruiting
+Fulvestrant NCT05646862 Breast cancer Phase III, Recruiting
+Palbociclib
+Fulvestrant
NCT04191499 Breast cancer Phase II/III, Recruiting
Serabelisib +Sapanisertib
+Paclitaxel
NCT03154294 Solid Tumors Phase I, Active, not recruiting
+Cisplatin
+Nab Paclitaxel
NCT03193853 Triple-negative breast cancer Phase II, Active, not recruiting
PI3Kγ Eganelisib +Etrumadenant
+PLD
NCT03719326 Triple-negative breast cancer Phase I, Completed
+Nivolumab NCT03980041 Urothelial carcinoma Phase II, Completed
+Atezolizumab
+Nab-paclitaxel/bevacizumab
NCT03961698 Triple-negative breast cancer,
Renal cell carcinoma
Phase II, Active, not recruiting
PI3Kδ Idelalisib +Lenalidomide NCT01838434 Mantle cell lymphoma Phase I, Completed
+Entospletinib NCT01796470 Chronic lymphocytic lymphoma,
Mantle cell lymphoma
Phase II, Terminated
+Obinutuzumab NCT03890289 Follicular lymphoma Phase II, Active, not recruiting
+Rituximab NCT01539512 Chronic lymphocytic lymphoma Phase III, Completed
+Bendamustine
+Rituximab
NCT01569295 Chronic lymphocytic lymphoma Phase III, Completed
+Ofatumumab NCT01659021 Chronic lymphocytic lymphoma Phase III, Terminated
PI3Kδ/CK1ε Umbralisib +Brentuximab vedotin NCT02164006 Hodgkin's lymphoma Phase I, Completed
+Ibrutinib NCT02268851 Chronic lymphocytic lymphoma,
Mantle cell lymphoma
Phase I, Active, not recruiting
+Ublituximab NCT02006485 Chronic lymphocytic lymphoma,
Non-Hodgkin lymphoma
Phase I, Completed
+Ublituximab
+Ibrutinib
NCT02006485 Chronic lymphocytic lymphoma,
Non-Hodgkin lymphoma
Phase I, Completed
+Rituximab NCT03919175 Follicular lymphoma,
Marginal zone lymphoma
Phase II, Recruiting
+Pembrolizumab NCT03283137 Non-Hodgkin lymphoma,
Chronic lymphocytic lymphoma
Phase I, Active, not recruiting
PI3Kδ Parsaclisib +Romidepsin NCT04774068 T-cell lymphoma Phase I, Recruiting
+CHOP NCT05238064 Peripheral T-cell lymphoma Phase I/II, Not yet recruiting
+Tafasitamab NCT04809467 Chronic lymphocytic lymphoma,
Non-Hodgkin lymphoma
Phase I/II, Active, not recruiting
+Itacitinib NCT04509700 B-cell malignancy Phase II, Recruiting
+Ibrutinib NCT04551066 Myelofibrosis Phase III, Recruiting
Zandelisib +Tazemetostat NCT05604417 Follicular lymphoma Phase I/II, Not yet recruiting
+Rituximab/Venetoclax NCT05209308 Chronic lymphocytic lymphoma Phase II, Withdrawn
+Rituximab NCT04745832 Follicular lymphoma,
Non-Hodgkin lymphoma
Phase III, Active, not recruiting
Linperlisib +Camrelizumab NCT05429398 Solid tumor Phase I, Not yet recruiting
+Azacitidine NCT05559008 Peripheral T-cell lymphoma Phase I/II, Recruiting
PI3Kα/δ Copanlisib +Durvalumab NCT04895579 Non-small cell lung cancer Phase I, Recruiting
+Trastuzumab NCT02705859 Breast cancer Phase I, Completed
+Venetoclax NCT04939272 Mantle cell lymphoma Phase I/II, Recruiting
+Nivolumab NCT03711058 Colorectal cancer Phase I/II, Active, not recruiting
+Avelumab NCT05687721 Bladder cancer Phase I/II, Not yet recruiting
+Rituximab NCT03474744 Marginal zone lymphoma Phase II, Recruiting
+Obinutuzumab NCT05387616 Follicular lymphoma Phase II, Recruiting
+R-CHOP NCT02626455 Non-Hodgkin lymphoma Phase III, Active, not recruiting
+Rituximab NCT02367040 Non-Hodgkin lymphoma Phase III, Active, not recruiting
Taselisib +Letrozole NCT02273973 Breast cancer Phase II, Completed
+Fulvestrant NCT02340221 Breast cancer Phase III, Terminated
PI3Kγ/δ Duvelisib +Romidepsin NCT02783625 T-cell lymphoma Phase I, Active, not recruiting
+Bortezomib NCT03534323 Chronic lymphocytic lymphoma Phase I/II, Recruiting
+Venetoclax NCT04688658 Melanoma Phase I/II, Recruiting
+Nivolumab NCT05057247 Head and neck squamous cell carcinoma Phase II, Recruiting
+Docetaxel NCT02576275 Non-Hodgkin lymphoma Phase III, Withdrawn
+Rituximab
+Bendamustine
NCT02204982 Follicular lymphoma Phase III, Terminated
Tenalisib +Pembrolizumab NCT03471351 Hodgkin lymphoma Phase I, Terminated
+Romidepsin NCT03770000 T-cell lymphoma Phase I/II, Completed
Pan-PI3K Buparlisib +Ibrutinib NCT02756247 Lymphoma Phase I, Completed
+Bevacizumab NCT01283048 Renal cell carcinoma Phase I, Completed
+Panitumumab NCT01591421 Colorectal cancer Phase I/II, Completed
+Bevacizumab NCT01349660 Glioblastoma Phase I/II, Completed
+Erlotinib NCT01487265 Non-small cell lung cancer Phase II, Completed
+Paclitaxel NCT01852292 Head and neck squamous cell carcinoma Phase II, Terminated
+Tamoxifen NCT02404844 Breast cancer Phase II, Completed
+Fulvestrant NCT01633060 Breast cancer Phase III, Terminated
+Paclitaxel NCT04338399 Head and neck squamous cell carcinoma Phase III, Recruiting
Pictilisib +Fulvestrant NCT01437566 Breast cancer Phase II, Completed
+Paclitaxel NCT01740336 Breast cancer Phase II, Completed
JAK1/2 Ruxolitinib +Venetoclax NCT03874052 Acute myeloid leukemia Phase I, Active, not recruiting
+Umbralisib NCT02493530 Myelofibrosis Phase I, Active, not recruiting
+Radiation;
+Radiation/temozolomide
NCT03514069 Glioma/Glioblastoma Phase I, Active, not recruiting
+Ibrutinib NCT02912754 Chronic lymphocytic lymphoma Phase I/II, Unknown
+Nilotinib NCT02973711 Chronic myelomonocytic leukemia Phase I/II, Withdrawn
+Nivolumab NCT03681561 Hodgkin lymphoma Phase I/II, Active, not recruiting
+Decitabine NCT02076191 Myeloproliferative neoplasm Phase I/II, Completed
+Trastuzumab NCT02066532 Breast cancer Phase I/II, Completed
+Erlotinib NCT02155465 Lung cancer Phase I/II, Completed
+Capecitabine NCT01423604 Pancreatic cancer Phase II, Completed
+Navitoclax NCT04468984 Myelofibrosis Phase III, Recruiting
Momelotinib +Capecitabine
+Capecitabine/oxaliplatin
NCT02244489 Pancreatic ductal adenocarcinoma Phase I, Terminated
+Erlotinib NCT02206763 Non-small cell lung cancer Phase I, Terminated
+Pembrolizumab NCT05436990 Melanoma Phase II, Not yet recruiting
+Durvalumab NCT04064190 Urothelial carcinoma Phase II, Not yet recruiting
+Durvalumab NCT04893252 Gastric cancer Phase II, Recruiting
JAK2 Fedratinib +Decitabine NCT05524857 Myeloproliferative neoplasm Phase I, Recruiting
+Ivosideni
+Enasidenib
NCT04955938 Myeloproliferative neoplasm Phase I, Recruiting
+Nivolumab NCT05393674 Myelofibrosis Phase II, Recruiting
Pacritinib +Ibrutinib NCT02677948 Chronic lymphocytic lymphoma, Small lymphocytic lymphoma Phase I/II, Withdrawn
+Decitabine
+Cytarabine
NCT02532010 Acute myeloid leukemia Phase II, Terminated
HPK1 CFI-402411 +Pembrolizumab NCT04521413 Advanced solid tumors Phase I/II, Recruiting
BGB-15025 +Tislelizumab NCT04649385 Advanced solid tumors Phase I, Recruiting
PF-07265028 +Sasanlimab NCT05233436 Advanced solid tumors Phase I, Recruiting
NDI-101150 +Pembrolizumab NCT05128487 Solid tumors Phase I/II, Recruiting
RORγt LYC-55716 +Pembrolizumab NCT03396497 Non-small cell lung cancer Phase I, Unknown
Bromodomain ZEN003694 +Nab-paclitaxel
+Pembrolizumab
NCT05422794 Metastatic triple-negative breast cancer Phase I, Recruiting
+Abemaciclib NCT05372640 Malignant solid neoplasm, NUT carcinoma Phase I, Suspended
+Talazoparib NCT05071937 Recurrent ovarian cancer Phase II, Recruiting
NCT03901469 Triple-negative breast cancer Phase II, Recruiting
NCT05327010 Malignant solid neoplasm, Ovarian carcinoma Phase II, Recruiting
+Binimetinib NCT05111561 Solid tumors Phase I, Recruiting
+Entinostat NCT05053971 Advanced/refractory solid tumors Phase I/II, Recruiting
+Etoposide
+Cisplatin
NCT05019716 NUT carcinoma Phase I/II, Recruiting
+Enzalutamide NCT04986423 Metastatic prostate cancer Phase II, Recruiting
NCT02711956 Metastatic prostate cancer Phase I/II, Completed
+Ipilimumab
+Nivolumab
NCT04840589 Solid tumors Phase I, Recruiting
+Enzalutamide
+Pembrolizumab
NCT04471974 Metastatic prostate cancer Phase II, Recruiting
GSK525762 +Trametinib NCT03266159 Solid tumors Phase II, Withdrawn
BI +Selumentinib
+Durvalumab
NCT05253131 Sarcoma Phase II, Not yet recruiting
Molibresib +Etoposide
+Cisplatin
NCT04116359 NUT carcinoma Phase I/II, Withdrawn
PLX51107 +Azacitidine NCT04022785 Myelodysplastic syndrome, Acute myeloid leukemia Phase I, Completed
SF1126 +Nivolumab NCT03059147 Advanced hepatocellular carcinoma Phase I, Terminated
GS-5829 +Exemestane
+Fulvestrant
NCT02983604 Advanced breast cancer Phase I/II, Terminated
Azacitidine +Pembrolizumab
+Epacadostat
NCT02959437 Advanced solid tumors Phase I/II, Terminated
OTX015 +Azacitidine NCT02303782 Acute myeloid leukemia Phase I/II, Withdrawn
DHODH JNJ-74856665 +AZA/VEN NCT04609826 Acute myeloid leukemia, Myelodysplastic syndromes, Chronic myelomonocytic leukemia Phase I, Active, not recruiting
Brequinar +Ribavirin NCT03760666 Acute myeloid leukemia Phase I/II, Terminated

2. Small-molecule agents targeting PRR-associated pathways

2.1. Targeting cGAS–STING pathway

Pattern recognition receptors (PRRs) play a central role in immune responses against various pathogen and damage-associated molecular patterns (PAMPs and DAMPs). Their activation elicits signaling cascades that lead to the initiation of cell-autonomous defense mechanisms, as well as the production of soluble mediators, such as type I interferon (IFN) and pro-inflammatory cytokines. By inducing the expression of IFN-stimulated genes, type I IFNs boost cell-autonomous defense mechanisms in an autocrine manner, and activate the adaptive immune system. Cytosolic DNA is a potent activator of a type I IFN response. Cyclic GMP–AMP synthase (cGAS)-stimulator of IFN genes (STING) axis is one of the most notable pathways that recognizes cytosolic DNA to drive activation of IFN and other inflammatory cytokines10. Activation of cGAS–STING in tumor cells may serve as a barrier to early neoplastic progression through the upregulation of inflammatory genes. Tumor DNA can also be released and transferred into the cytosol of dendritic cells (DCs) and macrophages. The accumulation of tumor DNA activates STING–IRF3-induced IFN signaling to enforce tumor-antigen presentation on DCs and macrophages that cross-prime CD8+ T cells for antitumor immunity (Fig. 1). The critical role of cGAS–STING signaling in antitumor immunity has sparked the development of pharmacologic agonists aiming to boost anti-tumor immunity or enhance the effects of existing immunotherapies. Over ten natural and synthetic STING agonists have undergone clinical development (Fig. 2A)11. Although generally not toxic, the first generation of STING agonists, cyclic dinucleotides (CDNs), are structurally unstable and only exhibit very modest activity via intratumoral delivery. The development of next-generation STING agonists, mainly novel CDNs and non-CDNs, with improved potency and properties enabling their stability for systemic delivery, represents the research direction in the field11, 12, 13. Extensive investigations are also invested to combine STING agonists with different classes of therapies in particular anti-PD-1/PD-L1 antibodies, as it has been shown that STING activation is accompanied by the upregulation of immune inhibitory factors including PD-L1.

Figure 1.

Figure 1

cGAS–STING signaling in antitumor immunity. Cytosolic DNA is a potent activator of type I IFN response. cGAS–STING pathway recognizes cytosolic DNA and induces the secretion of type I IFNs and other pro-inflammatory cytokines. Tumor cells are often replete with cytosolic DNA derived from genomic, mitochondrial, and exogenous origins. Activation of cGAS–STING in tumor cells may serve as a barrier to early neoplastic progression through the upregulation of inflammatory genes, which can recruit immune cells. Additionally, tumoral DNA is thought to be transferred and released into the cytosol of dendritic cells and macrophages. The accumulation of tumoral DNA, in turn, activates STING-induced IFN signaling to enforce tumor-antigen presentation on dendritic cells and macrophages that cross-prime CD8+ T cells for antitumor immunity. Created with BioRender.com.

Figure 2.

Figure 2

PRR-associated pathway agonists. (A) Representative STING agonists. ADU-S100, MK-1454, and TAK-676 are cyclic dinucleotides (CDNs) while SNX281 is non-CDN. (B) Representative TLR agonists. GSK1795091 is a TLR4 agonist and poly-ICLC (Hiltonol) is a TLR3 agonist, and the others are TLR7 agonists.

2.1.1. CDNs

The development of STING agonists was initially inspired by cyclic GMP–AMP (cGAMP), the endogenous ligand of STING. Modified CDNs are designed to mimic cGAMP. ADU-S100 was the first STING agonist to reach the clinical stage for cancer immunotherapeutic potentials. In its first-in-human trial, intratumoral administration of ADU-S100 demonstrated good tolerability among patients with advanced/metastatic solid tumors and lymphomas. However, its clinical efficacy was very modest, with only one confirmed partial response in Merkel cell carcinoma14. ADU-S100 was also investigated in combination with other ICIs, including spartalizumab and pembrolizumab, which was well tolerated but only minimal anti-tumor responses were observed14.

MK-1454 (Ulevostinag) is a rationally designed CDN based on the scaffold of 2′,3′-cGAMP, and ADU-S100. MK-1454 exhibited high affinity to human wild-type STING and its second-most prevalent variant (R71H-G230A-R293Q) with Kd values at single-digit nanomole or even lower. MK-1454 exhibited robust potency in cell-based assays and possessed a binding mode and kinetic binding profiles that closely resembled those of 2′,3′-cGAMP15. It is currently undergoing investigation in clinical trials (NCT03010176, NCT04220866), both as a monotherapy and in combination therapy.

2.1.2. Next-generation CDNs

To avoid the necessity for intratumoral delivery of early-generation CDNs, recent efforts have concentrated on developing STING agonists with stable physical properties suitable for systemic administration.

BMS-986301 is a next-generation CDN standing out for its promising preclinical results16. In a phase I trial targeting patients with advanced solid cancers (NCT03956680), BMS-986301 is evaluated either as monotherapy or in combination with nivolumab (Opdivo) and ipilimumab. SB11285 is a second-generation STING agonist that allows intravenous administration. The combination of SB11285 and cyclophosphamide was reported to produce a significant synergistic anti-tumor effect17. An ongoing phase I non-randomized, dose escalation study (NCT04096638) is currently evaluating SB11285 as monotherapy or in combination with atezolizumab for intravenous use in patients with advanced solid tumors. TAK-676 is another synthetic STING agonist designed for intravenous administration, which has been demonstrated to activate the STING signaling pathway and type I IFNs in a dose-dependent manner. TAK-676 exhibited remarkable effects in activating both innate and adaptive immune activity, as demonstrated in multiple syngeneic tumor models spanning various tumor types18. Currently, TAK-676 is being investigated in cancer patients with locally advanced or metastatic solid tumors, either as a monotherapy or in combination with various treatments including ICIs, radiation, and chemotherapy (NCT04420884, NCT04879849).

BI 1387446, a BI-STING compound, has progressed to clinical testing and is currently being evaluated in the first-in-human trial in patients with advanced, unresectable, and/or metastatic solid tumors (NCT04147234). The primary objective of this trial is to determine the maximum tolerated dose and assess the tolerability of a single intratumoral injection of BI 1387446, either as a monotherapy or in combination with BI 754091 (an anti-PD1 monoclonal antibody).

2.1.3. Non-CDNs

In addition to novel CDNs, an increasing number of small molecule non-CDNs STING agonists with drug-like physicochemical properties have been reported, including the first orally available STING agonist MSA-219 and a non-nucleotide cGAMP mimetic SR-71720, yet very few have entered the clinical stage.

GSK3745417 is a small molecule non-CDN with a dimeric amidobenzimidazole (ABZI) scaffold. This class of compounds was developed by linking two symmetry-related ABZI-based compounds to create linked ABZIs (diABZIs) to produce a synergistic antitumor effect. This novel dimeric design not only resolved the challenge of drug administration but also extended the therapeutic potentials, thus representing a milestone in the development of next-generation STING agonists20,21. Intravenous administration of GSK3745417 in syngeneic tumor models exhibited satisfactory plasma exposure and durable tumor regression. GSK3745417 is currently being investigated in a phase I dose-escalation study as monotherapy or in combination with dostarlimab in patients with relapsed/refractory solid tumors (NCT03843359, NCT05424380).

E7766 is a macrocycle-bridged STING agonist that structurally features a transannular macrocyclic bridge between the nucleic acid bases in CDNs, thereby effectively locking a bioactive U-shaped conformation. E7766 demonstrates enhanced stability and STING affinity due to the conformational rigidity provided by its unique macrocycle bridge, resulting in increased efficacy compared to conventional STING agonists. Furthermore, E7766 exhibits broad pan-genotypic activity across all major human STING variants. Currently, an ongoing phase I/Ib clinical trial (NCT04144140) is assessing the efficacy of intratumorally administered E7766 as a monotherapy in patients with advanced solid tumors and lymphomas22.

SNX281 is a small molecule STING agonist rationally designed by a multifaceted computational approach. SNX281 was created by using a unique self-dimerizing mechanism in the STING binding site, where the constituting ligands form dimers to resemble approximately the size and shape of a cyclic dinucleotide. Upon activation, it induces a large-scale conformational change of the STING protein. Preclinical data indicated that SNX281 exhibited adequate systemic bioavailability, and it was found to trigger STING-mediated cytokine release, strong induction of type I IFN, potent antitumor activity, durable immune memory, and single-dose tumor elimination in mouse models23. It is currently under investigation in a first-in-human clinical trial (NCT04609579) as a single agent or in combination with pembrolizumab for treating advanced solid tumors and lymphomas.

A few other non-CDN agonists are also currently under clinical investigation, though very limited information has been reported. IMSA101 is a small molecule analog of cGAMP. In preclinical studies, IMSA101 stimulated the production of IFNs and cytokines, generated long-term memory immunity to tumors, and demonstrated robust tumor growth inhibition as a single agent or in combination with anti-PD-L1 monoclonal antibody in multiple mouse models. A dose escalation (phase I) and dose expansion (phase IIa) study (NCT04020185) of intratumoral IMSA101 treatment alone or in combination with ICIs has been recruiting patients since 2019. MK-2118 is another non-CDN STING agonist whose structure has not been reported. In a clinical trial focusing on patients with advanced solid tumors or lymphomas (NCT03249792), SNX281 is being investigated, either intratumorally or subcutaneously injected as a monotherapy and in combination with pembrolizumab. KL340399 and HG381 are new-generation non-CDN STING agonists developed in China. They are currently under clinical investigation in phase I trials (NCT05549804, NCT04998422) to evaluate their safety, tolerability, pharmacokinetic profile, and antitumor efficacy in patients with advanced solid tumors.

2.1.4. STING agonists with facilitated delivery

Despite the substantial progress in designing novel cGAS–STING agonists, their biological efficacies are hindered by stability and delivery problems. Recently, novel delivery systems, including nanocarriers, microparticles, and hydrogels, have been developed to improve the pharmaceutical properties (e.g., excessive hydrophilicity, vulnerability to enzymatic degradation, and being negatively charged) of novel STING agonists.

Exosome-based formulation is believed to promote targeted antitumor immunity while minimizing off-target toxicity due to systemic elevation of toxic cytokines. CDK-002 is a novel exosome formulation loaded with an agonist of STING and it expresses high levels of glycoprotein prostaglandin F2 receptor negative regulator (PTGFRN). PTGFRN expression on the surface of the exosome facilitates specific uptake in tumor-resident antigen-presenting cells (APCs) and enhances systemic APC-mediated antitumor immune response. According to the information released by Codiak BioSciences, intratumoral administration of CDK-002 activates the innate immune response locally, thereby increasing the production of pro-inflammatory cytokines (including IFNs), enhancing the cross-presentation of tumor-associated antigens (TAAs) by DCs, and inducing a cytotoxic T-lymphocyte (CTL)-mediated immune response against cancer cells. A first-in-human, phase I/II open-label, multicenter, dose escalation, safety, pharmacodynamic, and pharmacokinetic study of CDK-002 (NCT04592484) has been initiated in patients with advanced/metastatic, recurrent, injectable solid tumors progressed following standard of care treatment.

2.2. Targeting Toll-like receptor pathways

Toll-like receptors (TLRs) family is one of the best-characterized PRRs responsible for sensing invading pathogens outside of the cell and in intracellular endosomes and lysosomes. Human TLR family consists of 10 receptors (TLR1-10), grouped into two major categories: surface TLRs (TLR1, TLR2, TLR4, TLR5, TLR6, and TLR10) that reside on the plasma membrane and bind to microbial-derived ligands, and intracellular endosomal TLRs (TLR3, TLR7, TLR8, and TLR9) that are nucleic acid-sensing TLRs. TLR4 is found both on the plasma membrane and in the intracellular compartments24, 25, 26. Responding to DNA/RNA derived from pathogens and dead cells, TLR signaling is activated to induce the production of pro-inflammatory cytokines or type I IFN and the activated TLRs expressed on DCs also contribute to T cell activation, thereby bridging between innate and adaptive immunity (Fig. 3).

Figure 3.

Figure 3

The roles of TLR in anti-infection and antitumor immunity. Innate immune cells (e.g., macrophages, dendritic cells, and NK cells) recognize the PAMP of microorganisms through PRRs such as TLRs. Macrophages kill bacteria and viruses via phagocytosis, whereas dendritic cells process the foreign antigens and present them to T cells through the TCR–MHC complex. Activated CD4+ T cells secrete cytokines to exert an immunoregulatory effect on CD8+ T cells and the latter differentiate into cytotoxic T cells to execute the cell-killing effect. PAMP, pathogen-associated molecular pattern; PRR, pattern recognition receptor; TLRs, Toll-like receptors. Created with BioRender.com.

Given the central role of TLRs in innate immunity and their important role in adaptive immunity, tremendous efforts have been invested in identifying TLR agonists to enhance antitumor immune response, particularly in the context of therapeutic vaccine potential in cancer. Different TLRs are amenable to the intervention via different types of agents (Fig. 2B). Cell surface TLRs can be targeted by small molecules and antibodies whereas the intracellular nucleic-acid sensing TLRs could be targeted by modified oligonucleotides. To date, agonists for several TLRs, including TLR3, TLR4, TLR7, TLR8, and TLR9, have been tested clinically either alone or in combination with other therapies to strengthen the immune system in cancer therapy24,27,28.

2.2.1. Intracellular TLR agonists

Imiquimod is an immune response modifier, which has been clinically approved for actinic keratosis, external genital warts, and basal cell carcinoma. It acts through a TLR7–MYD88-dependent pathway and has also been extensively investigated in a broad spectrum of cancer patients, mostly in combination with different types of cancer treatments, including tumor vaccines, chemotherapy, targeted therapies, and ICIs. Most recently, a phase I trial showed that the combination of topical imiquimod and IDH1 peptide vaccine resulted in immune responses in 93.3% of IDH1 R132H-mutated patients with grade I adverse effects29. In prostate cancer, the combination of topical imiquimod and different tumor vaccines also produced a promising antitumor effect30. Resiquimod (R848) and motolimod (VTX-2337) are second-generation derivatives of imiquimod. Resiquimod has been investigated in combination with tumor vaccine and/or other adjuvants for the treatment of melanoma and brain tumors, but it did not successfully induce a consistent antigen-specific CD8+ T cell response. Motolimod was tested in patients with ovarian cancer or other squamous cell carcinoma in different combination regimens. Among them, motolimod was shown to augment the clinical responses of patients with advanced head and neck squamous cell carcinoma (HNSCC) who received epidermal growth factor receptor inhibitor cetuximab31.

TLR9 is recognized as a first-line host defense against pathogens by recognizing DNA comprising unmethylated CpG motifs present in viruses and bacteria. CpG oligonucleotides (ODNs) can activate innate immunity by binding to TLR9 and trigger Th1-mediated immune responses. It has been hypothesized that the introduction of unmethylated CpG ODNs into tumor lesions may reinvigorate the tumor microenvironment (TME) and subsequently reverse therapeutic resistance to PD-1 blockade immunotherapy. Currently, the most extensively investigated TLR9 agonists in the clinic are SD-101, CMP-001, and IMO-2125. CMP-001 (Vidutolimod) is a virus-like particle containing a CpG-A TLR9 agonist. A recent phase Ib study (NCT02680184) showed that CMP-001 could reverse PD-1 blockade resistance by triggering a strong IFN response to induce and attract antitumor T cells. Among patients with advanced melanoma previously treated with pembrolizumab, 25% of patients had a partial (7 of 11) or complete (4 of 11) response to therapy, and responses were seen not only in the injected tumors but also in noninjected tumors, including visceral sites32,33. IMO-2125, also known as tilsotolimod, is a CpG-C TLR9 agonist that effectively activates DCs, induces type I IFN signaling, and upregulates multiple immune checkpoint pathways in cancer patients. In a phase Ib/II study, the combination of ipilimumab and IMO-2125 in 25 patients with advanced melanoma achieved a response rate of 36%34. However, a follow-up phase III trial evaluating the combination of ipilimumab with or without intralesional IMO-2125 did not improve the overall response rate (ORR), with 8.8% in the combination group versus 8.6% in ipilimumab alone group (NCT03445533).

2.2.2. Cell surface TLR agonists

TLR2 is a cell surface innate immune sensor that recognizes diverse ligands from external pathogens. TLR2 agonists, including triacylated lipopeptides, synthetic chemical compounds, glucomannan polysaccharides, naturally extracted compounds, and inactivated viruses, have been investigated to boost the antitumor response. In preclinical studies, TLR2 agonists alone or in combination with tumor vaccines, immune checkpoint inhibition, chemotherapy, photodynamic therapy (PDT), or adoptive cell transfusion, have been shown to activate immune responses. To date, there are only a limited number of clinical trials investigating TLR2 agonists for cancer immunotherapy. Amplivant, a synthetic TLR2 ligand that can be directly conjugated to tumor peptide antigens, represents the most promising one. In preclinical studies, amplivant-conjugation to antigens led to enhanced antigen presentation by DCs and T-cell priming, thereby inducing effective antitumor responses. Moreover, amplivant-conjugated synthetic long peptides (SLPs) have been shown to generate immune responses that are 100 times higher compared to unconjugated SLPs. In the first-in-human phase I clinical trial, amplivant-conjugated human papillomavirus (HPV) 16-SLP was administered as an intradermal therapeutic anticancer vaccine, and it was found to induce robust HPV16-specific T-cell immunity in patients with HPV16 positive malignancies35.

Among immune cells, TLR3 is expressed only in myeloid DCs, macrophages, and mast cells. TLR3 is known to localize both at the cell surface and in endosomes in mast cells and macrophages, but only in endosomes in myeloid DCs. TLR3 plays an important role in antiviral host response. Polyinosinic:polycytidylic acid (Poly (I:C)), a mismatched double-stranded RNA with one strand being a polymer of inosinic acid and the other a polymer of cytidylic acid, has been used as an immunostimulant. Poly (I:C) has been used in most immunotherapeutic studies as the TLR3-targeting agent because of its known interaction with TLR3. To date, three investigational TLR3 agonists, poly-ICLC (Hiltonol), rintatolimod (Ampligen), and BO-112 have progressed to clinical trials. Poly-ICLC is currently under active clinical investigation in combination with traditional therapies (surgery, chemotherapy, or radiotherapy) and immunotherapies (tumor vaccine, immune co-stimulation, or ICB) (NCT01976585, NCT03380871).

TLR4 is an important PRR that activates both innate and adaptive immune cells. TLR4 agonists under investigation for enhancing cancer immunotherapy include LPS, lipid A derivatives, polysaccharides, and protein TLR4 agonists. Monophosphoryl lipid A (MPLA) is the first TLR4 agonist approved as an adjuvant for hepatitis B vaccine. GLA-SE is a synthetic TLR4 agonist, which is composed of a glucopyranosyl lipid A-stable oil-in-water emulsion. The immune stimulatory activity of GLA-SE has been widely studied in lymphoma, skin cancers, sarcoma, lung cancer, and colorectal cancer. In a phase I trial (NCT02035657), intratumoral administration of G100 containing GLA-SE was well-tolerated as an adjuvant to surgery and radiotherapy, and it exhibited notable clinical efficacy in Merkel cell carcinoma with increased intratumoral infiltration of CD8+ and CD4+ T cells, activation of immune-related genes and local tumor regression36.

2.2.3. Other TLR agonists

In addition to single TLR activation, multi-TLR agonists are also under clinical investigation for cancer therapy. Bacillus Calmette-Guérin (BCG), an attenuated live Mycobacterium bovis simultaneously activating TLR2, TLR4, and TLR9, is the most studied multi-TLR agonist. Initially developed as a vaccine for tuberculosis, BCG has evolved to become a gold-standard adjuvant immunotherapy for patients with high-risk non-muscle-invasive bladder cancer. BCG was shown to induce a non-specific enhancement of the innate immune system, creating a heterologous immunological memory termed trained immunity37. Currently, BCG is being tested in bladder carcinoma patients in combination with other treatments, including anti-PD-1/PD-L1 antibodies, small molecule inhibitors, and a neoantigen-encoding gene vaccine. In a completed clinical trial (NCT02753309), the combination of rapamycin and BCG was well tolerated in patients with high-grade non-muscle invasive bladder carcinoma and it induced antigen-specific γδ T cell response as well as urinary cytokine production38. BCG was also investigated for the treatment of lower urinary tract carcinoma (NCT00794950). Besides malignancies in the urinary system, the combination of BCG and other cancer treatment modalities such as chemotherapy, radiofrequency ablation, and GM-CSF has also been investigated in the treatment of liver metastases from colorectal cancer (NCT04062721).

3. Small-molecule agents targeting immune checkpoints

3.1. Targeting PD-1/PD-L1 signaling

3.1.1. PD-1/PD-L1 signaling in immune-oncology

Programmed cell death protein 1 (PD-1) is an immunosuppressive molecule predominantly expressed by activated T cells and induces immunosuppressive signals through binding to its ligands, mainly programmed cell death-ligand 1 (PD-L1). The binding of PD-L1 to PD-1 triggers the phosphorylation of PD-1 and suppresses T cell receptor (TCR) and co-stimulatory signaling, resulting in the inhibition of T cell activation and function. Upregulation of PD-L1 expression mediated by cancer cells in TME fosters cancer immune escape, leading to limited antitumor responses of cytotoxic T cells. The blockade of the PD-1/PD-L1 signaling by ICIs represents an attractive strategy for cancer treatment. Numerous PD-1/PD-L1 ICIs exhibited potent antitumor efficacy by potentiating T cell-mediated antitumor immunity in clinical trials. However, currently clinically approved ICIs are monoclonal antibodies that have several disadvantages, such as poor tissue permeability, high rate of immune-related adverse events, and intravenous or subcutaneous routes of administration, thus hindering the widespread application of ICIs8. Conversely, small-molecule agents can overcome these drawbacks and exert more effective anticancer activity with fewer adverse effects.

In consideration of the intrinsic limitations of monoclonal antibodies, numerous small molecule inhibitors targeting PD-1/PD-L1 signaling were developed and some of them have entered clinical trials (Fig. 4)39. These small molecule agents inhibit PD-1/PD-L1-mediated immunosuppressive signal by preventing the binding of PD-1 to PD-L1, regulating PD-L1 expression, or various other mechanisms.

Figure 4.

Figure 4

Small-molecule immune checkpoint inhibitors. The PD-L1 inhibitors (INCB086550, GS-4224, IMMH-010, and MAX-10181) can interfere with the interaction of PD-1 and PD-L1, contributing to the inhibition of immunosuppressive signal. CA170 is a small-molecule inhibitor targeting both PD-L1 and VISTA.

3.1.2. Interfering with the interaction of PD-1 and PD-L1

INCB086550 is a small-molecule inhibitor targeting PD-L1 that potently contributed to PD-L1 dimerization and internalization, selectively interfered with the PD-1/PD-L1 interaction in vitro, and delayed tumor growth in CD34+ humanized mice40. Two clinical trials have been initiated to investigate INCB086550 in cancer patients (NCT03762447, NCT04629339).

IMMH-010 is an oral PD-L1 inhibitor, which could be rapidly metabolized to YPD-29B, and exhibited remarkable antitumor efficacy in xenograft mouse models of colon cancer and melanoma41. Currently, IMMH-010 is under clinical evaluation in advanced patients with solid tumors (NCT04343859). ASC61 is another small molecule PD-L1 inhibitor, which has received the investigational new drug approval for the treatment of advanced solid tumors. A phase I clinical study is underway to assess its safety (NCT05287399). GS-4224 (evixapodlin) was developed by Gilead Sciences for treating advanced solid tumors, however, the phase I study (NCT04049617) of GS-4224 was terminated for unknown reasons. Additionally, other PD-L1 inhibitors including MAX-10181 (NCT05196360, NCT04122339) and BPI-371153 (NCT05341557) are being studied in phase I clinical trials.

3.1.3. Regulating PD-L1 expression

The expression of PD-L1 is reported to be regulated by various mechanisms involved in transcriptional regulation, translational regulation, and epigenetic regulation. Multiple oncogenic signaling pathways consisting of JAK/STATs, RAS/RAF/MEK/ERK, PI3K/AKT/mTOR, and WNT/β-catenin were reported to have pivotal roles in regulating PD-L1 expression. PI3K/AKT/mTOR can increase PD-L1 expression through transcription factors including STAT3 and YAP1/TAZ, and mTOR-mediated translational regulation8. Activation of the AKT-mTOR pathway upregulated PD-L1 expression in NSCLC cells at the translational level, whereas rapamycin (mTOR inhibitor) inhibited PD-L1 expression by decreasing protein synthesis and increasing lysosomal protein degradation42. Furthermore, the combination therapy of rapamycin and anti-PD-1 markedly decreased tumor growth in a mouse model of lung cancer, with the increase of CD3+ T cells and reduction of FoxP3+ Tregs42. MAPK signaling can transcriptionally upregulate PD-L1 via AP1 and STAT3. EGF or IFNγ-induced activation of MAPK signaling increased the PD-L1 mRNA and protein, and selumetinib (MEK1/2 inhibitor) prevented the upregulation of PD-L1 in lung adenocarcinoma cells43. Small-molecule inhibitors targeting JAK–STATs (fedratinib, ruxolitinib, and tofacitinib) were also reported to downregulate PD-L1 expression in breast cancer cells and NSCLC cells44. Downregulation of PD-L1 expression through inhibition of oncogenic pathways could simultaneously suppress sustaining proliferative signaling and improve the immunosuppressive TME, contributing to enhanced antitumor efficacy.

In addition, epigenetic regulation also has an impact on PD-L1 expression. DNA global hypomethylation fosters PD-L1 expression while DNA hypermethylation within the PD-L1 promoter inhibits PD-L1 expression45. DNA hypomethylating agents (azacytidine and decitabine) could upregulate PD-L1 expression and potentiate the efficacy of anti-PD-L1 antibodies in several mouse models of cancer, such as NSCLC, gastric cancer, and colorectal cancer46,47. Histone acetylation at the PD-L1 promoter region also participates in the regulation of PD-L1 expression. Histone deacetylase (HDAC) inhibitors including belinostat, panobinostat, vorinostat, and romidepsin could increase PD-L1 expression and enhance the efficacy of anti-PD-1/PD-L1 antibodies in vivo48, 49, 50. Upregulation of PD-L1 mediated by epigenetic regulation potentially gives rise to enhanced antitumor efficacy of anti-PD-1/PD-L1 antibodies via increasing the proportion of cancer cells’ response to these antibodies. Currently, extensive clinical trials are underway to investigate the potential benefits of HDAC inhibitors combined with ICIs (NCT05068427, NCT03765229, and NCT04651127). Small-molecule drugs that regulate PD-L1 expression have many application prospects in cancer immunotherapy, however, it is worth noting that the mechanisms of action of these agents and options of drug combination rely on the cellular context and cancer types. Further clinical trials are warranted to broaden this promising research area.

3.2. Targeting VISTA signaling

V-domain immunoglobulin suppressor of T-cell activation (VISTA) is one of the immune checkpoint proteins that suppress T-cell response against cancer. VISTA, a member of the B7 protein family, shares 22% sequence similarity with PD-L151. The extracellular domain of VISTA contains two typical cysteines conserved in immunoglobulin-like proteins, and four unique cysteines only conserved in VISTA orthologs but absent in other B7 family members52. VISTA is abundantly expressed on myeloid and lymphoid cells53,54. The expression of VISTA on the γδ T cells and naïve CD4+ T cells blocks their auto-reactivation, thereby preventing T cell activation without foreign antigenic stimulation55. The binding of VISTA to its ligand can reduce the pro-inflammatory cytokines IL-2, TNF-α, and IFN-γ while increasing the anti-inflammatory cytokines and mediators. Deficiency or blockade of VISTA can greatly regulate the TME to a more pro-inflammatory myeloid phenotype, which is conducive to tumor-responsive T cell infiltration52,56,57. Therefore, VISTA-targeting therapy may potentiate the anticancer effect of immunotherapy57.

CA170 is an oral small-molecule antagonist targeting both PD-L1 and VISTA58. The drug molecule consists of l-serine, d-asparagine, and l-threonine, partially linked by diacylhydrazine and urea linker moieties. The in vitro inhibitory effect (EC50) of CA170 on PD-L1 and VISTA was found to be 66 nmol/L and 83 nmol/L, respectively. As a single agent, CA170 was taken orally once daily and significantly suppressed tumor growth and metastasis in mouse models of melanoma cells B16F10 and colorectal cells MC3859. CA170 was shown to rescue IFN-γ released from human PBMC blocked by the recombinant PD-L1, PD-L2 and VISTA. In a completed phase I study, CA170 was well tolerated at an oral dose of 50–1200 mg and it displayed a dose proportional plasma half-life of 4–9.5 h60. In a phase II study, CA-170 also demonstrated exciting clinical activity, including an ORR of 30% in Hodgkin lymphoma (based on Lugano criteria), and a clinical benefit rate of >85% at a daily dose of 400 mg and progression-free survival (PFS) of 19.6 weeks (PFS with best supportive care was approximately 8 weeks in a cross-study comparison) in advanced non-squamous NSCLC61. Notably, the development of lung cancer was completely inhibited by CA170 in combination with the major histocompatibility complex (MHC) class II-directed KRAS oncogene peptide vaccines. CA170 enhanced the tumor infiltration of CD8+ T cell and their effector functions by reducing the tumor infiltration of regulatory T (Treg) cells and myeloid-derived suppressor cells, and the KRAS vaccine mainly induced CD4+ effector T cells expansion62.

4. Small-molecule agents targeting cytokine and chemokine pathways

Cytokine and chemokine are important regulatory molecules in the TME and both play pleiotropic effects on cancer cells and immune cells. The cytokine and chemokine pathways are activated upon the binding of cytokine and chemokine to their corresponding receptors, which has an impact on tumor growth, proliferation, metastasis, and the function of immune cells. Thus, targeting immunoregulatory cytokines and chemokine contributes to enhanced antitumor immunity.

4.1. Targeting TGF signaling

TGF-β is a powerful regulatory cytokine that maintains immune homeostasis and tolerance via controlling lymphocyte proliferation, differentiation, and survival. TGF-β contains three isoforms. TGF-β1 is primarily involved in immunomodulation, whereas TGF-β2 and TGF-β3 play important roles in regulating cellular environments63. TGF-β1 and TGF-β3 are released from their latent complexes after the Arg–Gly–Asp motif interacts with integrins αvβ6 or αvβ8. The three isoforms of TGF-β are the ligands for the TGF-β receptor 1 (TβR1, ALK5) and TGF-β receptor 2 (TβR2) combination. TGF-β plays a critical dual role in the progression of cancer. In the early or primary stages of cancer, TGF-β can act as a tumor suppressor by inducing cell cycle arrest and promoting apoptosis. Cancer cells use TGF-β to initiate immune evasion, growth factor production, differentiation into an invasive phenotype, and metastatic dissemination, or establishment and expansion of metastatic colonies when cancer cells lose the tumor suppressor effect of TGF-β64. The direct effects of TGF-β on tumor cells and the indirect effects of TGF-β on tumor growth by creating a favorable microenvironment suggest that blocking TGF-β signaling is a beneficial response. Therefore, TGF-β inhibitors are expected to potentiate ICIs, especially in cancer types growing in TGF-β-rich environments. The development of small molecule inhibitors of TGF-β is an important complement to influence the tumor microenvironment. The initial TGF-β small-molecule inhibitor starts by targeting TβR1 (Fig. 5A)65.

Figure 5.

Figure 5

Cytokine and chemokine pathways inhibitors. (A) Chemical structures of representative TβR1 (ALK5) inhibitors. (B) Chemical structures of representative CSF1R inhibitors. (C) Chemical structures of representative chemokine receptor antagonists, including CCR2 antagonist (PF-04136309), CCR4 antagonist (FLX475), CCR5 antagonist (BMS-813160), CXCR1/2 antagonist (SX682), CXCR2 antagonist (AZD5069) and CXCR4 antagonist (LY2510924).

Galunisertib (LY2157299 monohydrate) is a potent small molecule inhibitor of TGFβR1 that specifically blocks the phosphorylation of SMAD2. Oral administration of 75 mg/kg twice daily of galunisertib was shown to significantly delay tumor growth of human lung cancer Calu6 xenograft model, human breast cancer MX1 xenograft model, and murine breast cancer 4T1 syngeneic model66. Galunisertib also exhibited good anticancer activity in preclinical models of myelodysplastic syndromes and acceptable side effects in phase I studies of solid tumors (NCT02008318)67. However, in a phase Ib clinical trial investigating the combination of galunisertib and ramucirumab (a VEGFR2 monoclonal antibody) to treat advanced hepatocellular carcinoma (HCC), the results did not endorse the preclinical hypothesis that inhibition of TGF-β signal transduction could enhance the efficacy of VEGF-targeted therapy (NCT01246986)68. In a phase IIa study investigating the combination of galunisertib and standard temozolomide-based radiochemotherapy (TMZ/RTX) to treat newly diagnosed malignant gliomas, the antitumor efficacy was found to be modest for the drug combination of galunisertib plus TMZ/RTX vs. TMZ/RTX (median overall survival (18.2 vs. 17.9 months), median PFS (7.6 vs. 11.5 months), and disease control rate (80% vs. 56%), respectively)69. In a recent phase Ib study for metastatic pancreatic cancer patients (NCT02734160), the combination of galunisertinib (150 mg twice daily) and durvalumab (anti-PD-L1 monoclonal antibody; 1500 mg Q4W) was well tolerated but clinical efficacy was limited70. The disease control rate for the drug combination was 25.0%. Median overall survival (OS) was 5.72 months, and PFS was 1.87 months. As the patients were not selected by using predictive biomarkers of TGF-β inhibition, the clinical benefit achieved by this study was limited70. In another phase II clinical trial, the combination of galunisertib and neoadjuvant chemoradiotherapy was well tolerated and was shown to increase the complete response rate to 32% in patients with locally advanced rectal cancer (NCT02688712)71, thus advocating further evaluation in larger randomized clinical trials. The results of a phase Ib/II study of galunisertib combined with nivolumab in NSCLC showed that the combination was well tolerated and observed in a subset of patients in stage II NSCLC to the initial effect (NCT02423343)72. In another phase II study of patients with recurrent glioblastoma, galunisertib in combination with lomustine failed to demonstrate an improvement in OS over lomustine alone (NCT01582269)73.

LY3200882 is an orally active ATP-competitive TGFβ-R1 inhibitor derived from galunisertib. It exhibited potent antitumor activity in a mouse model of triple-negative breast cancer, and its activity was associated with an increase in tumor-infiltrating lymphocytes. In immunosuppressive assays, LY3200882 was shown to rescue TGF-β1 inhibitory or T-modulating cells to inhibit naïve T cell activity and recover proliferation. In addition, LY3200882 also showed anti-metastatic activity in the intravenous mouse triple-negative breast cancer EMT6-LM2 model. Moreover, LY3200882 could also enhance the antitumor effect of checkpoint inhibition therapy (anti-PD-L1 monoclonal antibody) in the homologous mouse model of colorectal cancer74. A phase I multicenter clinical trial of oral LY3200882 was carried out, which included dose escalation, monotherapy extension for grade 4 gliomas, and combined therapy for solid tumors (LY3200882 plus PD-L1 inhibitor LY3300054), squamous cell carcinoma of head and neck (LY3200882, cis-platinum and radiotherapy), and pancreatic cancer (LY3200882, nab-paclitaxel and gemcitabine) (NCT02937272). Most (93.5%) of patients experienced an adverse event in ≥1 treatment, of which 39.6% were associated with LY320088275.

Vactosertib (TEW 7197) is another small-molecule inhibitor of TGFβ-R1. The combination of vactosertib and a PRMT5 inhibitor T1-44 could significantly reduce tumor size and invasion of surrounding tissues, and significantly improve long-term survival of the C57BL/6 mouse model of syngeneic orthotopic pancreatic ductal adenocarcinoma (PDAC)76. In other models including breast cancer, melanoma, and prostate cancer, vactosertib treatment abated cancer cell migration, invasion, and metastasis77, 78, 79. In a phase Ib/IIa clinical trial, the combination of vactosertib and pembrolizumab also showed promising antitumor efficacy and manageable safety in metastatic colorectal cancer patients who previously received chemotherapy80. Co-treatment of vactosertib with radiotherapy can enhance the treatment outcome of breast cancer by inhibiting epithelial–mesenchymal transition, reactive oxygen species stress generation, cancer cell stemness, and metastasis to other organs. Clinical trials of vactosertib plus other chemotherapies are being performed in patients with multiple cancer types, such as gastric cancer (NCT03698825), colorectal or gastric cancer (NCT03724851), desmoid tumor (NCT03802084), NSCLC (NCT03732274), and multiple myeloma (NCT03143985)81.

GFH018 is a small molecule inhibiting TGF-βR1 kinase. It is also a potent immune modulator by inhibiting Treg cell induction and reversal of the M2 phenotype to M1, thereby increasing the production of pro-inflammatory cytokines. GFH018 exhibits potent antitumor efficacy in several syngeneic mouse models as monotherapy or combinational therapy with anti-PD-L1 antibodies82. Currently, the combination of GFH018 and toripalimab is being investigated in patients with advanced solid tumors in an ongoing phase Ib/II clinical trial (NCT04914286).

AVID200 is a TGFβ1 trap that binds to TGFβ1 and TGFβ3, but not TGFβ2. AVID200 has been investigated in a phase I study (NCT03895112) which recruited 12 patients with myelofibrosis who were resistant, intolerant, or unsuitable for ruxolitinib and had grade 2+ myelofibrosis and platelet count >25 × 109/L. During dose escalation, 8 patients had grade 3/4 side effects, the majority of which were hematologic (anemia, thrombocytopenia), but dose-limiting toxicity (DLTs) did not occur. The spleen size was reduced by more than 50% in 2 patients, and the total symptom score improved by 50% in 5 patients. Interestingly, platelet counts improved in 8 patients, with a median increase of 48%83.

4.2. Targeting CSF1/CSF1R pathway

Colony stimulating factor 1 receptor (CSF1R) belongs to a member of the receptor tyrosine kinases family which is predominantly expressed in myeloid cells, such as DCs, macrophages, monocytes, microglia, and osteoclasts84. CSF1 activates CSF1R-mediated downstream signaling pathways and promotes the differentiation of hematopoietic stem/progenitor cells (HSPC) into heterogeneous populations of myeloid cells. However, abnormal activation of CSF1R signaling contributes to inflammatory diseases, neurodegenerative diseases, and cancer84, 85, 86. The CSF1/CSF1R pathway has attracted increasing attention for its roles in regulating the proliferation, migration, and survival of macrophages, and influencing immune function. Tumor-associated macrophages (TAMs) are recognized to exert immunosuppressive effects in the TME to promote tumor survival and metastasis. The function of CSF1R in modulating TAMs has been well-documented and pharmacological inhibition of CSF1R represents a novel therapeutic strategy for cancer therapy. CSF1R-targeted small-molecule inhibitors as monotherapy and combination treatment are currently in clinical development (Fig. 5B).

Pexidartinib (PLX3397) is an oral CSF1R inhibitor harboring activity against FLT3 and c-KIT, which received FDA approval for the treatment of adult patients with tenosynovial giant cell tumor (TGCT) based on positive results from the phase III ENLIVEN study87. In the ENLIVEN study recruiting 120 TGCT patients, pexidartinib achieved a higher ORR (39% vs. 0%) and improved patient symptoms compared with placebo88. However, severe adverse events were more common in the pexidartinib cohort than the placebo cohort (13% vs. 2%), and cholestatic hepatotoxicity was identified as a pexidartinib treatment-associated risk. CSF1R is primarily expressed in microglia in the central nervous system and it was reported that depletion of the microglia reduces tumor burden and invasive capacity. A phase II trial assessed the antitumor effect of pexidartinib in recurrent glioblastoma, however, the results showed no clinical efficacy from the pexidartinib monotherapy cohort89. The follow-up evaluation of the pexidartinib drug combination was suggested. Additionally, the safety and tolerability of pexidartinib plus sirolimus to target TAMs were evaluated in soft tissue sarcomas in a phase I study90. This combination therapy was well-tolerated, which supports further investigation to determine its clinical efficacy. Besides adult patients, pexidartinib was also well-tolerated in pediatric patients with leukemia and neurofibroma (NCT02390752). Moreover, pexidartinib monotherapy or combination treatment is being investigated in various cancers.

Surufatinib (sulfatinib, HMPL-012) is a kinase inhibitor that selectively targets CSF-1R, FGFR1, and VEGFR 1/2/3. The phase III SANET-ep trial investigated the efficacy of surufatinib among patients diagnosed with extrapancreatic neuroendocrine tumors (NETs)91. Surufatinib achieved a markedly longer PFS of 9.2 months vs. 3.8 months than placebo in advanced extrapancreatic NETs, with a favorable benefit-to-risk profile. Similarly, another phase III SANET-p study also showed that surufatinib could significantly prolong the median PFS (10.9 vs. 3.7 months) compared with a placebo for treating advanced pancreatic NETs92. Results from the two trials also revealed that the quality of life of these patients in the surufatinib group was similar to the placebo group except for diarrhea, which advocated the clinical application of surufatinib as a novel treatment option in such a patient population. Based on the promising results of these trials, surufatinib received its first approval in China for treating extrapancreatic NETs in 2020. Surufatinib as either monotherapy or combinatorial therapy is currently under investigation in patients with thyroid cancer, ovarian cancer, breast cancer, and other types of solid tumors (Table 1, Table 2).

Chiauranib is a novel multitargeted inhibitor that simultaneously inhibits CSF1R, VEGFR1-3, PDGFRα, and c-Kit, which has activity against tumor growth, angiogenesis, and chronic inflammation. In preclinical studies, chiauranib presented antitumor potential in mouse models of multiple cancers, including colorectal cancer, non-Hodgkin lymphoma (NHL), acute myeloid leukemia (AML) and HCC. A phase I trial revealed that chiauranib demonstrated favorable pharmacokinetic traits and an acceptable safety profile with potential antitumor effect among patients with refractory advanced solid tumors93. Chiauranib monotherapy has entered phase III assessment for patients with small cell lung cancer (SCLC) after two lines of chemotherapy (NCT04830813). Additionally, chiauranib is also under active clinical investigation in a range of cancers as monotherapy or combination therapy.

Compared with pexidartinib, vimseltinib (DCC-3014) has been shown to exhibit significantly improved selectivity to CSF1R and more durable suppression of CSF1R activity in vitro and in vivo. Vimseltinib was also found to significantly suppress tumor growth in mouse models of tumors. Vimseltinib is currently being studied as monotherapy in TGCT and combined with avelumab (anti-PD-L1 antibody) for sarcoma in various ongoing clinical trials at different stages (NCT03069469, NCT05059262, NCT04242238).

In a phase I/II trial (NCT02880371), while combination therapy of the CSF1R-selective inhibitor ARRY-382 (PF-07265804) with pembrolizumab was well-tolerated, it only exhibited limited clinical benefit in cancer patients, which resulted in premature termination of the study94. Similarly, another selective inhibitor of CSF-1R, JNJ-40346527 (edicotinib) also showed limited clinical activity against refractory or relapsed Hodgkin lymphoma in phase I/II trial95. Additionally, BLZ945 (sotuletinib) alone and combination therapy with spartalizumab (anti-PD-1 antibody) are underway to be assessed in advanced solid tumors (NCT02829723).

Collectively, CSF1R inhibitors represent a novel category of immunomodulatory drugs, however, the clinical efficacy is limited because of the existence of other immunosuppressive molecules and cells in TME. The combination of CSF1R inhibitors with chemotherapy, anti-angiogenic drugs, or immunotherapy may provide better clinical outcomes and they are under clinical evaluation.

4.3. Targeting chemokine receptors

Chemokines and their receptors are important mediators of immune cell trafficking, which play a central role in the composition of TME, proliferation, and metastasis of cancer cells96. Leukocyte recruitment and activation, angiogenesis, cancer cell proliferation, and metastasis are regulated by chemokines and their receptors. The precise movement of immune cells, such as leukocyte recruitment, was controlled by the spatial and temporal expression of chemokines. Chemokines can directly stimulate the growth of cancer cells via activating various signaling pathways, including PI3K/AKT/NF-κB and MAPK/ERK pathways. Importantly, the chemokine/chemokine receptor pathways have pleiotropic effects on tumor cells and immune cells. Some exert antitumor function by inducing the activation and differentiation of immunostimulatory cells while others suppress antitumor immune response (Fig. 6). There have been many studies involving chemokines and their receptors which could be used as therapeutic targets in solid tumors. Numerous chemokines/chemokine receptors-targeted drug candidates that focus on hematological or solid tumors are under clinical trials as monotherapy or in combination with anti-PD1/PD-L1 (Fig. 5C). Two antagonists, plerixafor and maraviroc, have been approved for clinical use. Other chemokine receptor antagonists including cenicriviroc, balixafortide, and motixafortide are in various phases of clinical trials.

Figure 6.

Figure 6

Chemokines and chemokine receptors in the tumor microenvironment. Chemokines are known to play dual roles in the TME. Some chemokines recruit T cells and NK cells to exert antitumor immunity, whereas other chemokines recruit Treg and MDSCs to impair immune activity. The effect of chemokines on diverse immune cell populations reflects their complex biological function. Created with BioRender.com.

4.3.1. CCR2 antagonists

The chemokine CCL2 is an important mediator of chemotaxis secreted by myeloid cells in tumors. The CCL2/CCR2 signaling axis promotes tumor cell proliferation and invasiveness, and recruits immunosuppressive cells97. Thus, cancer treatment has been increasingly concerned with blocking or inhibiting the CCL2/CCR2 signaling axis.

PF-04136309 (INCB8761) is a potent, selective, and orally bioavailable CCR2 antagonist. Researchers previously found that when PF-04136309 was combined with FOLFIRINOX chemotherapy, the partial response rate was higher (49%), as compared to FOLFIRINOX chemotherapy alone in patients with locally advanced (PDAC). Interestingly, the clinical benefit achieved by PF-04136309 was associated with the accumulation of CCR2+ inflammatory monocytes in the bone marrow98. On the other hand, the combination of PF-04136309 and nab-paclitaxel plus gemcitabine was well tolerated in patients with metastatic PDAC, but without significant clinical benefit in another phase Ib trial (NCT02732938)99.

CCX872, also known as CCX872-B, is a potent and orally active CCR2 antagonist. It improved the efficacy of the anti-PD-1 treatment in a preclinical setting. Combination therapy of CCX872 with FOLFIRINOX leads to enhanced tumor response in locally advanced PDAC patients100.

As a small molecule CCR2/5 dual antagonist, BMS-813160 is now in phase I/II trials (NCT03496662, NCT03767582)101.

4.3.2. CCR4 antagonists

The CC chemokine receptor 4 (CCR4) is important for regulating immune balance and it is mainly expressed in Th2 and Treg cells, which respond to CCL17 and CCL22102. Recent studies demonstrated that CCR4 blockade could limit the immunosuppressive functions of Tregs, thus making it a promising strategy for enhancing cancer immunotherapy. While numerous preclinical studies supported the emerging role of CCR4 as an immunotherapy target, there were only limited clinical studies conducted in this area in solid tumors.

FLX475 is a highly potent CCR4 antagonist that blocks the recruitment of Tregs to tumors. A phase II clinical trial has been initiated to evaluate the safety and antitumor activity of FLX475 alone or in combination with ipilimumab in subjects with advanced melanoma (NCT04894994). This trial was recently terminated due to the changes in the clinical landscape and no data have been released. Another two clinical studies (NCT04768686, NCT03674567) are underway to investigate the combination of FLX475 and pembrolizumab in patients with advanced cancer including gastric cancer.

4.3.3. CCR5 antagonists

The CC chemokine receptor 5 (CCR5) plays a key role in the recruitment of immune cells to sites of inflammation by directing immune cell migration along the chemokine gradient. It is activated by several chemokine ligands, including CCL3, CCL4, CCL5 and CCL8. In cancer biology, the CCR5 signaling pathway is primarily involved in immune surveillance, and it promotes tumor growth, stimulates angiogenesis, and suppresses tumor metabolic reprogramming and tumor stem cell proliferation103.

CCR5 antagonists, including maraviroc and vicriviroc, combined with pembrolizumab in patients with microsatellite stable (MSS) CRC are currently under clinical investigation (NCT03274804, NCT03631407)104,105, which showed that the OS was extended when maraviroc combined with pembrolizumab in such group of patients.

BMS-813160 is a CCR2/CCR5 dual antagonist with potential immunomodulatory and antineoplastic activities. A phase II trial is currently ongoing to evaluate the efficacy of BMS-813160 (CCR2/5-inhibitor) or BMS-986253 (anti-IL-8 antibody) in combination with nivolumab for NSCLC or HCC (NCT04123379)106. The combination of BMS-813160 and nivolumab is also being evaluated for patients with PDAC, microsatellite-stable colon cancer, and primary liver cancer (NCT04123379)101. Meanwhile, a phase I/II trial of neoadjuvant and adjuvant nivolumab and BMS-813160 with or without the GVAX vaccine is also under active investigation for locally advanced PDAC (NCT03767582).

4.3.4. CXCR1/2 antagonists

The chemokine receptors 1 and 2 (CXCR1/2) play a crucial role in angiogenesis, inflammation, chemotaxis of inhibitory myeloid cells, and tumor cell survival. As one of the major CXCR1/2 ligands, IL-8 (CXCL8) was overexpressed in solid tumors to promote tumor growth. Importantly, high serum IL-8 level was found to correlate well with poor response to ICIs. Therefore, inhibition of the IL-8/IL-8 receptor axis has been proposed as an emerging strategy to potentiate ICI efficacy. A series of clinical trials are underway to evaluate the safety and efficacy of combination therapy of anti-PD-1 antibody and IL-8 receptor antagonist, including SX682 (CXCR1/2 antagonist), navarixin (CXCR1/2 antagonist) and AZD5069 (CXCR2 antagonist) (NCT04599140, NCT03473925, and NCT02499328) in patients with advanced/metastatic solid tumors. Additionally, reparixin is an investigational allosteric inhibitor of CXCR1/2. The combination of reparixin and paclitaxel was shown to be safe, with demonstrated responses, in patients with HER2-negative metastatic breast cancer (NCT02001974)107.

4.3.5. CXCR4 antagonists

CXCR4, a chemokine receptor, is widely expressed in immune response cells. It modulates various cellular functions, including cell migration, chemotaxis, differentiation, proliferation, and apoptosis, thus contributing to its vital role in both cancer development and progression108,109. A CXCR4 antagonist, plerixafor, has received FDA approval for autologous transplantation in lymphoma and multiple myeloma patients110.

A few other novel CXCR4 antagonists have also been developed for cancer therapy. LY2510924 is a potent CXCR4 antagonist under investigation in clinical studies. The combination of LY2510924 and sunitinib was tolerated but it did not improve the efficacy of sunitinib alone in patients with metastatic renal cell carcinoma (NCT01391130) in a phase II study111. Recently, the maximum tolerated dose (MTD), safety, and tolerability of the combination of LY2510924 and durvalumab were also evaluated in a phase Ia study in patients with advanced (metastatic and/or unresectable) solid tumors. The recommended dose showed acceptable safety and tolerability112.

BL-8040 (motixafortide) is a novel selective inhibitor of the CXCR4 chemokine receptor. A multi-center phase IIa trial of BL-8040 in combination with pembrolizumab and chemotherapy was completed. Compared with pembrolizumab, BL-8040 increased CD8+ effector T cell tumor infiltration in pancreatic cancer patients. The study revealed that CXCR4 blockade gave rise to clinical efficacy and safety benefits in metastatic PDAC (NCT02826486)113.

5. Small-molecule agents targeting metabolic pathways

The accelerated proliferation of tumor cells in the TME demands a substantial supply of nutrition, resulting in the shortage of nutrients and accumulation of metabolites. The depletion of nutrients and increased harmful metabolites will impair immune cell differentiation and their normal physiological function, collectively disrupting antitumor immunity. Several key enzymes that modulate important metabolic pathways in the TME have been investigated in clinical trials. Some of the representative metabolic pathway inhibitors are summarized in Fig. 7.

Figure 7.

Figure 7

Metabolic pathway inhibitors. (A) Representative arginase inhibitors. Arginase inhibitors include boronic acid derivatives (numidargistat) and arginine homologs (nor-NOHA). (B) Representative IDO1 inhibitors. (C) Representative prostaglandin pathway inhibitors. Prostaglandin pathway inhibitors are comprised of COX-2 inhibitors (celecoxib) and EP4 antagonists (ONO-4578, E7046 and CR6086). (D) Representative adenosine pathway inhibitors.

5.1. Targeting arginine metabolism

Arginase, an enzyme containing manganese, carries out the last step of the urea cycle, which aims to eliminate harmful ammonia from the body. This process involves converting l-arginine into l-ornithine and urea. It has two isoforms, namely ARG1 and ARG2114. Increased expression of arginase may deplete arginine, which is associated with malfunction of T cells. Reduced concentration of arginine could suppress the mTORC1 activity of T cells and impair their normal function in immune response115. Furthermore, arginine depletion can lead to the arrest of T cell cycle progression at the G0–G1 phase, a decrease in IFN-γ levels, and disruption of TCR signaling116. Moreover, the depletion of arginine could also activate the NF-κB-GCN2/eIF2α pathway in lymphocytes and suppress its protein synthesis and proliferation117. Importantly, it has been shown that supplementation of arginine could rescue the proliferation and anti-tumor response of T cells118. The underlying mechanism of the immunosuppressive role of arginine metabolism is summarized in Fig. 8.

Figure 8.

Figure 8

Overexpression of ARG1 and ARG2 leads to arginine depletion and T-cell dysfunction. The arginases ARG1 and ARG2 are expressed by MDSCs and DCs/tumor cells, respectively. The overexpression of ARG1 and ARG2 in TME results in arginine depletion, which could lead to T-cell dysfunction. Arginine depletion can suppress the mTORC1 activity of T cells, arrest cell cycle progression of T cells at the G0–G1 phase, diminish the production of IFN-γ, and disrupt TCR signaling. MDSCs, myeloid-derived suppressor cells; DCs, dendritic cells; TME, tumor microenvironment. Created with BioRender.com.

In the TME, ARG1 is mainly expressed in myeloid-derived suppressor cells (MDSCs)119. It has been reported that the overexpression of ARG1 in MDSCs can effectively suppress the responses of T cells (CD4+ and CD8+) and NK cells and create an immunosuppressive microenvironment that promotes tumor growth in PDAC patients120. Furthermore, research has demonstrated that inhibiting ARG1 expression in MDSCs can effectively restore T cell function and enhance the antitumor immune response against multiple myeloma116. In ovarian cancer patients, extracellular vehicles (EVs) containing ARG1 were detected in the plasma and ascites121. Tumor-secreted EVs were additionally discovered in the lymph nodes, where they were observed to hinder the proliferation of T cells. Significantly, when these patients were administered arginase inhibitors as part of their treatment, the population of CD4+ and CD8+ T cells was restored, and the growth of tumors was slower. This represents the initial report documenting the transfer of a metabolic checkpoint molecule through tumor-derived extracellular vesicles over a long distance.

The upregulation of ARG2 has been observed in cancer cells, T cells, and DCs. This overexpression of ARG2 was identified in various types of cancer, resulting in arginine depletion within the TME. Consequently, the normal functions of T cells are suppressed122. Moreover, the overexpression of ARG2 in T cells was known to retard their cell proliferation and thus leading to immunosuppression123. Conversely, downregulation of ARG2 expression in T cells could restore their antitumor activity, though the extracellular level of arginine was not significantly altered. On the other hand, the antitumor response from DCs was also impaired by ARG2 overexpression124. To this end, DCs are known to produce microRNA-155 (miR155) to activate the antitumor effect of T cells. When ARG2 is overexpressed in DCs, the expression of miR155 is suppressed to mediate cell cycle arrest of T cells.

Considering the critical role of arginase in immunosuppression, numerous arginase inhibitors have been designed and investigated to enhance the outcome of cancer immunotherapy by promoting the proliferation of T cells and NK cells. The most established arginase inhibitors include arginine homologs (e.g., nor-NOHA) and boronic acid derivatives (e.g., numidargistat). Numidargistat (CB-1158, INCB01158) demonstrated the ability to inhibit MDSC-mediated immunosuppression of T cell proliferation in vitro, both as a standalone treatment and in combination with other cancer therapies124. To date, two clinical trials of phase I/II have been initiated to test the effectiveness of numidargistat as a monotherapy and in combination with pembrolizumab, an anti-PD-1 monoclonal antibody in the treatment of various cancer types (NCT02903914; NCT03910530).

5.2. Targeting indoleamine 2,3-dioxygenase 1 (IDO1)

l-Tryptophan (Trp) is an essential amino acid that is exclusively acquired through the diet. Trp metabolism is involved in the regulation of immunity and its aberrant regulation could cause cancer. IDOs (indoleamine 2,3-dioxygenases), as well as TDO (tryptophan 2,3-dioxygenase), are enzymes involved in the process of tryptophan deoxygenation, converting tryptophan into kynurenine. IDO1 is the most fully characterized and it is overexpressed in the vast majority of cancers125. Functionally, IDO1 plays a pivotal role in cancer immune escape via depleting tryptophan in the TME, which leads to T-cell anergy and apoptosis via the General Control Nonderepressible 2 (GCN2) pathway. In addition, the accumulation of the downstream products of IDO1 including kynurenine and quinolinic acid could induce the differentiation of naïve CD4+ T cells towards the immunosuppressive form of T cells (CD3+CD4+CD25+FOXP3+ Treg) (Fig. 9). On the other hand, IDO1 overexpression could cause immunosuppression in a non-enzymatic manner mediated by MDSCs. Furthermore, IDO1 overexpression is known to cause resistance to immune checkpoint inhibitors. For example, overexpression of CTLA-4 by Treg could cause the upregulation of IDO1 expression in DCs126. PD-1 expression in T cells can be upregulated by IDO1 overexpression through aryl hydrocarbon receptor activation127. When both CTLA-4 and PD-1 expression were inhibited in glioblastoma-bearing mice, the expression of IDO1 was found upregulated128. A comprehensive analysis of data from The Cancer Genome Atlas (TCGA) demonstrated a strong correlation between the expression of IDO1 and several other immune checkpoints including PD-L1, PD-L2, CD39, and FoxP3129. Since the overexpression of IDO1 is often correlated with other immune checkpoints, IDO1 inhibitor was often used in combination with other cancer immunotherapy to achieve better treatment outcomes.

Figure 9.

Figure 9

The role of IDO1 in immunosuppression. IDO1 is expressed in tumor cells, MDSCs, CAF, and TAM. Overexpression of IDO1 leads to the depletion of tryptophan in the TME, thereby triggering the activation of GCN2 signaling, inhibiting mTORC1 activity, and retarding cell cycle progression of T cells. Meanwhile, the accumulation of kynurenine, a metabolite produced by IDO1, can induce the generation of Treg cells and contribute to immunosuppression mediated by MDSCs and TAM. MDSCs, myeloid-derived suppressor cells; CAF, cancer-associated fibroblasts; TAM, tumor-associated macrophage. Created with BioRender.com.

Early phase I/II clinical trials suggested that administering an IDO1 inhibitor as a standalone treatment showed limited effectiveness in cancer therapy. Nonetheless, clinical studies have shown that combining an IDO1 inhibitor with other cancer therapies produced a synergistic effect. This was evidenced by enhanced suppression of tumor growth and prolonged survival of patients130. Indoximod (D-1-MT), epacadostat (INCB024360), IDO1 vaccines, and other IDO1 inhibitors have progressed to various phases of clinical trials130, 131, 132. In a phase II clinical trial (NCT02178722), the combined therapy of epacadostat, an IDO1 inhibitor, and the anti-PD-1 monoclonal antibody pembrolizumab exhibited promising efficacy in treating various types of solid tumors. Unfortunately, the combination of epacadostat and pembrolizumab did not show a significant improvement in antitumor efficacy in phase III clinical trials conducted in patients with metastatic melanoma (NCT02752074) or urothelial carcinoma (NCT03361865).

5.3. Targeting prostaglandin pathway

Cyclooxygenase (COX) is the key enzyme catalyzing the biosynthesis of prostaglandins (PGs), which are important mediators of inflammation in various diseases. Two isoforms of COX enzymes, COX-1 and COX-2, have been identified. COX-2 is an established target of nonsteroidal anti-inflammatory drugs (NSAIDs) which also represents a novel molecular target for cancer therapy. Previous research has reported that constitutive IDO1 expression is driven by COX2 and mediates intrinsic immune resistance in many cancer types. Prostaglandin E2 (PGE2) is the primary metabolite of arachidonic acid generated by COX-2 and it regulates many biological functions including inflammatory activation and triggers a range of cellular responses through binding to one or more of its four prostaglandin E (EP) receptors (EP1, EP2, EP3, and EP4). In addition, EP receptors are also coupled to heterotrimeric G proteins which initiate intracellular signaling cascades, such as MAPK, NF-κB, glycogen synthase kinase 3β (GSK3β)-β catenin, Src–EGFR–Akt–mTOR, and EP4–PKA–CREB pathways, thereby facilitating tumor progression133.

Importantly, COX-2/PGE2 signaling is recognized for its significant contribution in shaping an immunosuppressive TME134. COX-2 is released into the TME by cancer-associated fibroblasts (CAFs), alternatively activated macrophage type 2 (M2) cells, as well as tumor cells. Kabir et al.135 showed that COX-2 and PGE2 were overexpressed in senescent CAFs, which contributed to tumor progression. The release of COX-2 from M2 cells favors tumor angiogenesis, invasion, and metastasis136, 137, 138. In tumor cells, the activation of MAPK, EGF, and KRAS signals, as well as inflammation and hypoxia, can upregulate COX-2 expression by upregulation of PTGS2 (Fig. 10)139. COX-2 overexpression and over-activity can induce upregulation of the immunosuppressor cells including MDSCs, and TAMs, thus the manipulation of which could be exploited to enhance or suppress immune responses in cancer140. Tumor-derived PGE2 is known to diminish the viability and chemokine production of NK cells, which is accompanied by down-regulating chemokine receptor expression in type 1 DCs (cDC1)141. Moreover, tumor production of PGE2 acts selectively on EP2 and EP4 receptors in NK cells, subsequently preventing the switch of inflammatory TME from cold to hot, and promoting immune evasion142. These studies demonstrate the potential of targeting PG pathways to improve immune checkpoint blockade efficacy against cancer.

Figure 10.

Figure 10

The role of COX-2/PGE2 signaling in promoting immunosuppressive tumor microenvironment. Hypoxia, inflammation, and intrinsic factors within the tumor contribute to the upregulation of COX-2 and the excessive production of PGE2 in tumor cells, thus creating an immunosuppressive tumor microenvironment through diverse mechanisms. COX-2/PGE2 signaling is associated with the accumulation of MDSCs and M2 polarization of macrophages. Additionally, the COX-2/PGE2 signaling pathway suppresses the recruitment of DCs and hampers the activation of NK cells and T cells. MDSCs, myeloid-derived suppressor cells; DCs, dendritic cells. Created with BioRender.com.

5.3.1. COX-2 inhibitors

Celecoxib is a potent and selective COX-2 inhibitor approved by the FDA in 1998 for treating familial adenomatous polyposis. It has also been investigated for its effect on immunity and metabolism in the TME in different cancer types in clinical trials, mainly as adjuvant treatment combined with other checkpoint inhibitors. However, clinical outcomes from these drug combinations were mostly unsatisfactory. In a recent phase III trial, the effects of adding celecoxib to standard adjuvant chemotherapy for 3 years, compared to a placebo, did not yield a significant improvement in disease-free survival for patients with resected stage III colon cancer. Another phase III trial also showed no significant clinical benefit after a two-year treatment period with celecoxib compared to placebo as adjuvant treatment of HER2-negative breast cancer143,144. A few other ongoing clinical trials are investigating the combination of celecoxib and other treatment modalities. A phase I/II study is trying to confirm whether neoadjuvant PD-1 blockade with toripalimab, with or without the addition of celecoxib, could potentially serve as a more effective therapeutic approach for patients with mismatch repair deficient or microsatellite instability-high, locally advanced colorectal cancer145.

5.3.2. EP4 antagonist

As PGE2 regulates immune cell function, the modulation of EP receptor signaling pathways has emerged as attractive targets to enhance cancer immunotherapy. Among the four EP receptors, the EP4 receptor has been identified as a particularly useful cancer target. In patients with advanced or metastatic solid tumors, ONO-4578, a highly selective EP4 antagonist, exhibited potent antitumor activity and it was well tolerated when used in combination with nivolumab146. In a preclinical study, ONO4578 demonstrated potent antitumor activity and it was also shown to reduce infiltration of M2 macrophages in tumor xenografts in experimental mice. A phase I study is ongoing to research the combination of nivolumab with ONO-4578 in advanced or metastatic solid tumors (NCT03155061).

E7046 (AN0025), a novel EP4 receptor-specific antagonist, was found to exhibit manageable tolerability, immunomodulatory properties, and durable disease control (≥18 weeks) in patients with advanced malignancies147. A phase Ib, open-label, multicenter study (NCT03152370) of E7046 in combination with radiotherapy or chemoradiotherapy in patients with rectal cancer has been conducted recently, but the findings have not been released. There is another ongoing phase I trial that investigates the combination of E7046 and atezolizumab in locally advanced solid tumors.

CR6086 is another potent EP4 antagonist, which displays considerable immunomodulation activity. A phase I/Ib study is currently underway to evaluate the efficacy of CR6086 in combination with balstilimab (PD-1 inhibitor), focusing on patients with pretreated mismatch-repair-proficient and microsatellite stable metastatic colorectal cancer (NCT05205330).

5.4. Targeting adenosine signaling

Adenosine signaling derived from the A2a receptor (A2aR) triggers the cAMP/PKA/CREB pathway through typical G proteins, which is emerging as an important checkpoint of the immune response. Adenosine signaling is mediated via G-protein coupled adenosine receptors (A1R, A2aR, A2bR, A3R) and modulates human immunity. Adenosine is mainly produced by ectonucleotidases (CD39 and CD73) dephosphorylating extracellular ATP. It has been reported that the adenosine pathway was activated in the tumor microenvironment and inhibited the function of CD8+ T cells, natural killer (NK) cells, and macrophages (Fig. 11)148. Besides, adenosine stimulates DCs to secrete immunosuppressive factors like IL-10, TGFβ, arginase, and IDO149.

Figure 11.

Figure 11

The activation of adenosine signaling pathway in the tumor microenvironment. Adenosine signal inhibits the function of CD4+ T cells, CD8+ T cells, and NK cells by reducing the production of immune factors including IL-2 and IFN-γ. Adenosine also promotes the secretion of immunosuppressive factors such as IL-13 and IL-14. Therefore, the activation of the adenosine signaling pathway in TME promotes the formation of an immunosuppressive TME. Created with BioRender.com.

The A2a receptor (A2aR) is a typical GPCR with a high affinity for adenosine. It is expressed on T cells, B cells, natural killer T (NKT) cells, monocytes, macrophages, DCs, and NK cells. When the A2aR signaling pathway is activated in CD4+ T cells, less IL-2 or IFN-γ is secreted, and CD28 is suppressed in these cells. Adenosine signaling can be inhibited by targeting CD73 and CD39 in TME, as well as blocking A2a and A2b receptors to enhance the anti-tumor immune response. In combination with other immune checkpoint inhibitors such as PD-L1, TIM-3, or CTLA-4, A2aR antagonists have been shown to potentiate anti-tumor immunity150. To this end, some small-molecule A2aR antagonists including ciforadenant, imaradenant, etrumadenant, and taminadenant have been developed.

Ciforadenant (CPI-444), a potent, selective, oral A2aR antagonist, alone or in combination with anti-PD-1/PD-L1 was shown to potently restrain tumor growth in multiple preclinical tumor models151,152. In clinical studies, ciforadenant combined with anti-PD-1/PD-L1 can increase the number of CD8+ T cells in tumor tissues of patients with renal cell carcinoma (RCC), including patients who had progressed on PD-(L)1 inhibitors153. Additionally, ciforadenant plus CPI-006 in adult subjects with advanced cancers is in progress in phase I/Ib open-label trial (NCT03454451). Among patients with metastatic castration-resistant prostate cancer, colorectal cancer, or relapsed/refractory solid tumors, another A2aR antagonist, imaradenant (AZD4635) monotherapy (75–200 mg twice daily or 125 mg once daily) and in combination with anti-PD-1/PD-L1 inhibitors exhibited good tolerance154.

Etrumadenant, also known as AB928, is a selective A2aR and A2bR dual antagonist developed by Arcus Company. When combined with docetaxel or zimberelimab, etrumadenant was shown to produce clinical benefits for patients with metastatic castration-resistant prostate cancer155,156. Furthermore, updated results from the ARC-3 study showed that the combination of etrumadenant and modified FOLFOX chemotherapy was well tolerated and associated with a substantial disease control rate in metastatic colorectal cancer patients157.

Last but not least, taminadenant (NIR178; PBF509), a potent selective A2aR antagonist, combined with spartalizumab (PDR001) was well tolerated in patients with advanced NSCLC. However, no clinical benefit was observed in this study (NCT02403193)158.

6. Small-molecule agents targeting oncogenic pathways

It has long been known that oncogenic pathways provide sustaining proliferative signaling for cancer cells and reprogram cellular metabolism, contributing to their rapid growth. In addition, it is reported that oncogenic pathways also play a crucial role in immunomodulation, which influences the biological function of immune cells and shapes the immunosuppressive environment.

6.1. Targeting PI3K-mediated signaling

The phosphatidylinositol 3-kinase (PI3K) family can be divided into class I, class II, and class III PI3Ks based on their structure and mode of regulation. The class I PI3Ks are comprised of PI3Kα, PI3Kβ, PI3Kγ, and PI3Kδ, and each of them contains a regulatory subunit and a catalytic subunit. PI3Kα and PI3Kβ are present in a broad spectrum of cell types, whereas PI3Kγ and PI3Kδ are predominantly enriched in leukocytes. The PI3K pathway is often abnormally activated in cancer and contributes to tumor initiation and progression. Additionally, the PI3K pathway is also implicated in the development, differentiation, and activation of lymphocytes, regulating the biological function of the immune system159. Hyperactive PI3Kδ signaling induced by gain-of-function mutations in PIK3CD and PIK3R1 leads to activated PI3Kδ syndrome (APDS), a primary combined immunodeficiency syndrome160. It is well known that macrophages play dual roles in inflammation and cancer, and an interesting study demonstrated that PI3Kγ signaling controls the molecular switch between immunostimulatory and immunosuppressive transcriptional programs in macrophages via AKT and mTOR161. Furthermore, oncogenic mutations in PIK3CA (encoding PI3Kα) have pleiotropic effects in solid tumors. On the one hand, activating PIK3CA mutations provides sustaining proliferative signaling for cancer cells; on the other hand, paracrine effects induced by PIK3CA mutations in cancer cells perhaps contribute to immune suppression through secretion of cytokines and chemokines and depletion of metabolic nutrients in the stroma162, 163, 164. Owing to the important role of PI3K signaling in immunity and cancer, inhibition of class I PI3Ks becomes a hotspot of drug development. Besides targeting tumor cell-intrinsic PI3K activity, increasing evidence underlines the potential of PI3K inhibitors in cancer immunotherapy (Fig. 12)165. PI3K inhibitors, particularly PI3Kα and PI3Kδ inhibitors, have achieved great progress in recent years and more than a dozen of PI3K inhibitors have entered clinical trials, which could be employed to reshape the TME and enhance antitumor immunity (Fig. 13A)166. Importantly, several drugs have received FDA approval for the clinical application of solid tumors and hematologic malignancies.

Figure 12.

Figure 12

The role of PI3K inhibition in antitumor immunity. PI3K signaling not only provides the sustaining proliferative signaling for tumor cells but also has an important role in the regulation of immune components in the TME. Thus, PI3K inhibition can suppress tumor growth and inhibit the tumor-promoting function of immunosuppressive cells, contributing to enhanced antitumor immunity. Created with BioRender.com.

Figure 13.

Figure 13

Oncogenic pathway inhibitors. (A) Representative PI3K inhibitors. PI3K inhibitors consist of PI3Kα inhibitors (alpelisib, inavolisib and serabelisib), PI3Kδ inhibitors (idelalisib, umbralisib, parsaclisib, zandelisib and linperlisib), PI3Kγ inhibitor (eganelisib), dual PI3K inhibitors (copanlisib, taselisib, duvelisib and tenalisib) and pan-PI3K inhibitors (buparlisib and pictilisib). (B) Representative JAK inhibitors. JAK inhibitors include ruxolitinib, fedratinib, momelotinib, and pacritinib.

6.1.1. PI3Kα inhibitors

Alpelisib is a potent PI3Kα-selective inhibitor and the first clinically approved PI3Kα inhibitor. PIK3CA activation mutations are frequent in breast cancer and they could be used as a predictive biomarker for anticancer activity of PI3Kα inhibitors. In HER2-negative, estrogen receptor (ER)-positive breast cancer, PIK3CA mutations occur in approximately 40% of cases. It was reported that PI3Kα-selective inhibitors enhance ER function and dependence on estrogen in ER-positive breast cancer167. Therefore, hormone-responsive breast cancer is particularly responsive to the antitumor effect of PI3Kα inhibitors.

The phase III SOLAR-1 trial compared the effect of alpelisib plus fulvestrant (ER antagonist) with placebo plus fulvestrant in HER2-negative, ER-positive, PIK3CA-mutated, advanced breast cancer patients who had received prior endocrine therapy. The alpelisib–fulvestrant group achieved a longer PFS (11.0 vs. 5.7 months) than the placebo group, which subsequently led to FDA approval of the drug combination168. According to the final results from the SOLAR-1 study, the median OS was 39.3 months and 31.4 months in the combination group and placebo group, respectively169. Furthermore, alpelisib–fulvestrant combination was also found to significantly prolong median OS (37.2 vs. 22.8 months) in patients with lung and/or liver metastases.

The success of alpelisib fosters the development of other PI3Kα-selective inhibitors. Inavolisib is another recently developed PI3Kα-selective inhibitor that induces the selective degradation of PI3K mutant p110α protein (encoded by PIK3CA) and has 300-fold selectivity over the other class I PI3K isoforms170. This unique mechanism of action of inavolisib has little effect on wild-type p110α protein, which retains normal physiological function mediated by PI3K signaling. In early-phase clinical trials, inavolisib demonstrated antitumor efficacy with an acceptable safety profile in PIK3CA-mutated breast cancer patients. The antitumor efficacy of inavolisib in combination with fulvestrant and alpelisib combined with fulvestrant is studied in an ongoing clinical trial of PIK3CA-mutated, HER2-negative, hormone receptor (HR)-positive, locally advanced or metastatic breast cancer patients (NCT05646862). Besides, the combinations of inavolisib with other anticancer agents are also under active clinical investigation in other solid tumors (NCT04449874, NCT03006172, NCT04929223, and NCT04931342).

Serabelisib (TAK-117/MLN1117/INK1117) is another selective PI3Kα inhibitor. While serabelisib as single-agent therapy was shown to produce only limited efficacy in a phase I study, combination therapies involving serabelisib were evaluated171. Mechanistically, the simultaneous inhibition of PI3K and mTOR may produce more remarkable inhibition on the PI3K/AKT/mTOR pathway than the individual PI3K or mTOR inhibition alone. Serabelisib combined with sapanisertib (mTORC1/2 inhibitor) was reported to produce a synergistic antitumor effect in the preclinical model of bladder cancer. However, in a completed phase II trial (NCT02725268), the dual PI3K/mTOR inhibition by sapanisertib and serabelisib failed to prolong PFS (3.1 vs. 3.6 months) compared with sapanisertib alone in advanced RCC patients after antiangiogenic therapy172. The three-drug combination treatment (serabelisib plus sapanisertib and paclitaxel) is being evaluated in patients with advanced solid tumors. In a phase I trial (NCT03154294), such a regimen presented preliminary remarkable efficacy and manageable safety profile, with a PFS of 11 months.

6.1.2. PI3Kδ inhibitors

Idelalisib (GS-1101/CAL-101) is a selective inhibitor of the PI3Kδ subunit, which was reported to inhibit B cell receptor (BCR) signaling in malignant B cells. It is the first PI3Kδ inhibitor approved for treating relapsed follicular lymphoma (FL) and chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL) based on promising results of idelalisib monotherapy and combination therapy with rituximab173,174. Idelalisib monotherapy showed a PFS of 11 months and a response rate of 57% in indolent NHL patients, and neutropenia is the most common ≥grade 3 adverse event. Idelalisib plus rituximab (anti-CD20 antibody) significantly improved PFS (19.4 months vs. 6.5 months) and OS (40.6 months vs. 34.6 months) compared with rituximab alone in relapsed CLL, with a favorable safety profile175. Furthermore, the addition of idelalisib to bendamustine (chemotherapeutic drug) and rituximab achieved a longer PFS of 20.8 months vs. 11.1 months than bendamustine combined with rituximab in relapsed or refractory CLL patients176. However, serious adverse events were more frequent (68% vs. 44%) in the idelalisib group than in the placebo group, and an increased risk of infection (39% vs. 25%) was observed in the idelalisib group. Similarly, idelalisib plus the second-generation anti-CD20 antibody ofatumumab also exhibited a better PFS of 16.3 months vs. 8.0 months than the ofatumumab group in patients with relapsed CLL177. The exciting results obtained from these clinical trials supported the clinical application of idelalisib in combination with an anti-CD20 antibody for treating relapsed CLL.

Umbralisib (TGR-1202) is another approved PI3Kδ inhibitor for treating relapsed or refractory marginal zone lymphoma (MZL), CLL, and FL adult patients. The safety and efficacy of umbralisib monotherapy in lymphoma and leukemia were assessed in several clinical trials (NCT01767766, NCT02793583, and NCT02742090), which demonstrated its superior clinical efficacy and a relatively low incidence of side effects. Given that the combination treatment of PI3Kδ inhibitor with anti-CD20 antibody exerts a potent antitumor effect in B-cell malignancies, umbralisib combined with CD20-directed monoclonal antibody was also studied. In a phase 1/1b study (NCT02535286), the promising clinical efficacy of umbralisib plus ublituximab (anti-CD20 antibody) showed that the median duration of response (DOR) was 20 months and ORR was 46%178. Additionally, it was reported that both PI3Kδ and Bruton tyrosine kinase (BTK) are implicated in the BCR pathway, thus, dual PI3Kδ and BTK blockade could be an efficacious antitumor strategy159,160. Another phase I trial (NCT02268851) demonstrated umbralisib plus ibrutinib (BTK inhibitor) was well-tolerated in B-cell malignancies, suggesting BTKi and PI3Kδi doublet treatment should be assessed in further investigations. In addition, the safety and efficacy of umbralisib, ublituximab, and ibrutinib combination therapy are being studied in an ongoing phase I trial (NCT02006485), which will provide better insight into this triplet combination in the management of CLL and NHL179.

Parsaclisib (INCB50465) is a highly selective PI3Kδ inhibitor investigated in multiple studies of B cell malignancies (NCT02018861, NCT02998476, and NCT04509700). Parsaclisib monotherapy or combined with chemotherapy or JAK1 inhibitor for the treatment of relapsed or refractory B-cell malignancies was assessed in a phase I/II study. Parsaclisib monotherapy showed antitumor activity in relapsed or refractory B-cell NHL with ORR of 30% in diffuse large B-cell lymphoma (DLBCL), 67% in mantle cell lymphoma (MCL), 71% in FL and 78% in MZL180. Additionally, parsaclisib was also underway in a range of solid tumors in combination with different agents including immunotherapy and targeted therapy (NCT02559492, NCT04551066).

Other PI3Kδ inhibitors including zandelisib (ME-401), linperlisib (YY-20394), AMG319, and IOA-244 are in different stages of clinical investigation. Zandelisib as monotherapy or combined with rituximab is currently investigated in a phase I trial of relapsed or refractory B-cell malignancies (NCT02914938). Linperlisib showed promising preliminary efficacy with ORR of 64.0% and 79.8% in a phase I (NCT03757000) and a phase II (NCT04370405) study, respectively181,182.

6.1.3. PI3Kγ inhibitors

Compared with PI3Kδ inhibitors, fewer drug candidates were developed to target PI3Kγ, and no PI3Kγ-selective inhibitors have received regulatory approval for treating cancer patients, indicating the distinct biological functions of PI3Kγ and PI3Kδ in tumors. Kaneda et al.161 demonstrated that PI3Kγ promotes immune suppression through the production of immune-suppressive macrophages (M2) in the TME. Eganelisib (IPI-549), the only PI3Kγ-selective inhibitor studied in clinical trials, can reprogram the TME and foster the transformation of M2 into antitumor macrophages (M1)183. Additionally, eganelisib overcame resistance to immune checkpoint blockade (ICB) among patients with high infiltration of immunosuppressive myeloid cells by targeting PI3Kγ in myeloid cells184. Notably, eganelisib synergizes with anti-PD1 to promote tumor regression and improve survival in mouse models. Based on these preclinical studies, eganelisib monotherapy or in combination with ICB or chemotherapy are currently under clinical investigation in different cancer types (NCT03795610, NCT02637531, and NCT03961698).

6.1.4. Dual PI3K inhibitors

Besides selective inhibitors targeting individual PI3K isoforms, dual PI3K inhibitors have also been developed. Simultaneous inhibition of PI3Kδ and PI3Kγ was reported to enhance antitumor cytotoxicity by promoting human CAR-T cell epigenetic and metabolic reprogramming185,186. Moreover, the combination of PI3Kδ/γ inhibition and irradiation was also found to potentiate effector CD8+ T cell-dependent antitumor immunity185,186. Therefore, dual PI3K inhibitors are expected to give rise to more potent inhibition of PI3K signaling and they represent promising antitumor therapeutic strategies.

Copanlisib is a pan-class I PI3K inhibitor that exhibits predominant activity against the p110α and p110δ isoforms and is also considered a PI3Kα/δ inhibitor. Dreyling et al.187 demonstrated that copanlisib monotherapy achieved ORR of 43.7% vs. 27.1% and median PFS of 294 days vs. 70 days in the indolent lymphoma and the aggressive lymphoma, respectively. Similarly, the median PFS was 11.2 months and the ORR was 59% in indolent lymphoma patients who received copanlisib after two or more lines of therapy. Furthermore, gene expression analysis indicated that the high response rate of copanlisib was related to the high expression of PI3K and BCR signaling genes188. The two studies both demonstrated the antitumor activity and well-tolerated safety profile of copanlisib monotherapy in indolent lymphoma. On account of the promising results of these studies, copanlisib received the FDA accelerated approval for the treatment of relapsed FL adult patients who have been previously treated with at least two systemic therapies. Combination therapy of copanlisib and gemcitabine was also an effective treatment option with ORR of 72% and PFS of 6.9 months in relapsed/refractory peripheral T cell lymphoma patients189. Notably, a phase III study investigated copanlisib combined with rituximab vs. placebo in combination with rituximab in relapsed indolent NHL patients. Copanlisib plus rituximab showed a more substantial clinical benefit as reflected by a significantly higher median PFS (21.5 months vs. 13.8 months) than the placebo group. However, it is noteworthy that more serious adverse events (47% vs. 18%) were reported in the combination group than in the placebo group, and the most frequent grade 3–4 adverse events were hyperglycemia (56% vs. 8%) and hypertension (40% vs. 9%)190.

Taselisib is a potent dual PI3Kα/δ inhibitor. Similar to inavolisib, taselisib was also known to promote the degradation of the mutated p110α isoform. The early-stage trials suggested that the combination therapy of taselisib and estrogen receptor (ER) antagonist (e.g., fulvestrant and tamoxifen) had preliminary evidence of antitumor activity in ER-positive advanced or metastatic breast cancer (NCT02285179, NCT01296555). On account of these studies, the phase III SANDPIPER study was conducted to assess the clinical efficacy of taselisib combined with fulvestrant. The combination treatment of taselisib and fulvestrant was found to improve PFS (7.4 vs. 5.4 months) compared with the placebo cohort in PIK3CA-mutant, HER2-negative, ER-positive advanced breast cancer. However, such a combination regimen has limited clinical utility because of modest clinical benefit and considerable serious adverse events, resulting in the termination of its clinical development191. Similar results were also observed in other PIK3CA-mutant cancers including NSCLC, head and neck squamous, and cervical cancer.

Duvelisib is a dual PI3Kγ/δ inhibitor that was clinically approved for treating adult patients with relapsed or refractory FL, CLL, and SLL. In the phase II DYNAMO trial (NCT01882803), duvelisib monotherapy exhibited clinically meaningful activity with a median PFS of 9.5 months and an ORR of 47.3% in patients with refractory indolent NHL. Furthermore, in the phase III DUO study, compared to ofatumumab, duvelisib demonstrated a longer PFS of 13.3 months vs. 9.9 months and a better ORR of 74% vs. 45% in relapsed or refractory SLL/CLL patients (NCT02004522)192. Apart from duvelisib monotherapy, combination treatments are also under active clinical investigations in a range of cancer types. Duvelisib plus rituximab or bendamustine/rituximab achieved a median PFS of 13.7 months and an ORR of 71.8% in NHL or CLL patients, and the clinical efficacy of such regimen should be further studied.

Tenalisib (RP6530) is another dual PI3Kγ/δ inhibitor. Given that the PI3Kγ and PI3Kδ isoforms are overexpressed in Hodgkin lymphoma cells within the TME, the antitumor activity of tenalisib against Hodgkin lymphoma was investigated in vitro and in vivo. Tenalisib inhibited the proliferation of Hodgkin lymphoma cells and transformed macrophages from an M2-like phenotype to an M1-like state, resulting in tumor regression193. Currently, tenalisib monotherapy and its combination with romidepsin are being investigated in several clinical trials (NCT03711578, NCT05021900, and NCT03770000).

6.1.5. Pan-PI3K inhibitors

Activation of the PI3K pathway is known to confer resistance of hormone receptor (HR)-positive breast cancer to endocrine therapy. Thus, a combination treatment of PI3K inhibitor and endocrine therapy could be a feasible and efficacious option for HER2-negative, HR-positive advanced breast cancer. Buparlisib, a pan-PI3K inhibitor, was shown to potently inhibit all class I PI3K. Buparlisib plus fulvestrant presented a better median PFS of 6.9 months vs. 5.0 months and 3.9 months vs. 1.8 months than placebo plus fulvestrant in such patients group in the BELLE-2 and BELLE-3 trials, respectively (NCT01610284, NCT01633060). However, safety concerns for the buparlisib–fulvestrant combination in both trials, due to the low selectivity of buparlisib to PI3K inhibition, have halted its further clinical development. Moreover, the combination of buparlisib and paclitaxel also failed to improve PFS (9.1 vs. 9.2 months) compared to paclitaxel plus placebo in HER2-negative breast cancer, whereas this combination improved PFS (4.6 vs. 3.5 months) in head and neck squamous cell carcinoma (HNSCC)194,195. Thus, buparlisib plus paclitaxel is being investigated in a phase III trial for the treatment of HNSCC (NCT04338399). Similar to buparlisib, another pan-PI3K inhibitor pictilisib combined with paclitaxel or fulvestrant did not improve PFS (6.6 months vs. 5.1 months and 8.2 months vs. 7.8 months) of HR-positive breast cancer, respectively (NCT01437566, NCT01740336). Collectively, these clinical studies indicate that pan-PI3K inhibitors exert limited antitumor efficacy with toxicity related to their non-selective PI3K inhibition.

6.2. Targeting JAK-mediated signaling

The Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway plays a critical role in inflammation, hematopoiesis, adipogenesis, tissue repair, and immune responses. Multiple cytokines, growth factors, and hormones exert biological function through the JAK–STAT pathway. However, the aberrant activation of JAK/STAT signaling contributes to inflammatory and immune-mediated diseases, as well as malignancies196,197. For example, elevated IL-6 produced in the TME leads to hyperactivation of JAK/STAT3 signaling, resulting in the proliferation, invasiveness, and metastasis of cancer cells. In addition, IL-6-mediated JAK/STAT3 also exerts negative regulatory effects on DCs, NK cells, and effector T cells, but positively regulates Treg cells and MDSCs, collectively promoting an immunosuppressive TME198. In consideration of the regulatory function of JAK/STAT signaling in the immune system and its tumor-promoting effect in tumor development, targeting the JAK/STAT pathway has emerged as a useful therapeutic regimen in cancer treatment. Currently, over a dozen JAK inhibitors are under different stages of clinical development, and some of them have been approved for the treatment of a heterogeneous group of disorders, including autoimmune diseases and hematological malignancies199,200. Clinical applications of many JAK inhibitors mainly focus on inflammatory and autoimmune diseases, including ulcerative colitis, systemic lupus erythematosus, and rheumatoid arthritis199. Several JAK inhibitors (ruxolitinib, fedratinib, momelotinib, and pacritinib) have received FDA approval for treating myeloproliferative neoplasms (MPN) on account of promising results in clinical trials (Fig. 13B)201, 202, 203. In this section, we mainly focus on the JAK inhibitors involved in MPN. Myelofibrosis is a kind of MPN associated with the activation of the JAK2-mediated signaling pathway.

Ruxolitinib is a selective and potent JAK1/2 inhibitor that showed clinically significant activity against myelofibrosis in early-stage clinical studies. The phase III COMFORT-I and COMFORT-II studies demonstrated that ruxolitinib exhibited substantial clinical benefits in patients with myelofibrosis, contributing to its accelerated drug approval201,204. In the COMFORT-I trial, 41.9% of patients in the ruxolitinib group achieved a reduction in spleen volume ≥35% at 6 months compared to 0.7% in the placebo group. More patients treated with ruxolitinib (45.9% vs. 5.3%) showed an improvement in myelofibrosis-related symptoms at 24 weeks, and significant OS benefit was observed in the ruxolitinib group. Similarly, the favorable antitumor efficacy of ruxolitinib was also observed in the COMFORT-II trial, which evaluated the safety and efficacy of ruxolitinib in patients with myelofibrosis compared to the best available therapy (BAT). Ruxolitinib showed a marked and durable reduction in splenomegaly with a decrease of 56% in length and improved symptoms associated with myelofibrosis. Moreover, the addition of navitoclax (Bcl-2 inhibitor) to ruxolitinib also resulted in improvement in spleen volume, hemoglobin, and bone marrow fibrosis205. Besides myelofibrosis, ruxolitinib monotherapy and in combination with chemotherapy, immunotherapy or targeted therapy are under active clinical investigations in different clinical stages for a range of cancers, including breast cancer, lung cancer, and pancreatic cancer.

Fedratinib is a potent JAK2-selective inhibitor approved for first-line and second-line therapy of myelofibrosis. The JAKARTA study demonstrated that 400 mg and 500 mg fedratinib were more effective than placebo in patients with myelofibrosis, which substantially decreased splenomegaly and symptom burden202. The phase II JAKARTA-2 trial investigated the efficacy of fedratinib in patients with myelofibrosis who received ruxolitinib, indicating that 55% of patients achieved spleen responses206. However, it is noteworthy that fedratinib treatment was related to an increased risk of Wernicke's encephalopathy, thus, considerations should be taken for dose adjustment and more closely monitoring.

Pacritinib is another approved JAK2 inhibitor for myelofibrosis. Pacritinib is focused on AML and myelofibrosis patients because of its high selectivity against the JAK2 V617F and FLT3 D835Y mutants frequently observed in AML and MPN. The two PERSIST-1 and PERSIST-2 studies compared the clinical efficacy of pacritinib with BAT in myelofibrosis203,207. Pacritinib exhibited superior efficacy to BAT in terms of symptom improvement and spleen volume reduction. Additionally, pacritinib 200 mg twice daily is more effective than pacritinib 400 mg once daily in improving hemoglobin and reducing transfusion burden. Pacritinib has also been evaluated in several other types of cancers, including breast cancer, prostate cancer, and colorectal cancer.

Momelotinib, a potent JAK1/2 inhibitor, was evaluated in two phase III trials for the treatment of myelofibrosis208,209. The SIMPLIFY-1 study assessed the safety and efficacy of momelotinib compared to ruxolitinib in myelofibrosis patients who never received JAK inhibitors. Momelotinib was non-inferior to ruxolitinib in terms of spleen response, and the results demonstrated that 26.5% of patients in the momelotinib cohort and 29% of patients in the ruxolitinib cohort achieved ≥35% spleen volume reduction. However, the SIMPLIFY 2 trial suggested that momelotinib was not superior to BAT (mainly ruxolitinib) in myelofibrosis patients previously receiving ruxolitinib for the reduction of spleen volume by ≥ 35%. Anemia is a common symptom of myelofibrosis and momelotinib can improve anemia with reductions in symptoms and splenomegaly. Thus, the phase III study (MOMENTUM) investigated the clinical benefits of momelotinib in patients with anemia and myelofibrosis. Momelotinib was shown to produce clinically significant improvements in spleen response, anemia, and myelofibrosis-associated symptoms than the traditional danazol treatment, which supported momelotinib as an effective treatment option in patients with myelofibrosis, especially in those with anemia210. Moreover, the exciting clinical outcome also led to FDA approval of momelotinib for patients with anemia and myelofibrosis in the first-line and second-line settings.

Taken together, JAK inhibitors generally exhibit clinical efficacy for the treatment of myelofibrosis. However, given the complexity of myeloproliferative neoplasms, JAK inhibitor monotherapy appears inadequate and its combination with other anticancer modalities should be developed. With the accumulating knowledge of JAK–STAT pathway in tumor biology, more JAK inhibitors will be developed and their full potential in cancer therapy could be realized in the future.

7. Small-molecule agents targeting immune-related kinases

7.1. Targeting hematopoietic progenitor kinase 1 (HPK1)

A member of the MAP4Ks family of human STE20-like protein serine/threonine kinases is the hematopoietic progenitor cell kinase 1 (HPK1), also known as mitogen-activated protein kinase kinase kinase kinase 1 (MAP4K1)211. Numerous studies have demonstrated that HPK1, which functions as a negative regulator of T cell, B cell, and DC-mediated immune responses, collaborates with several signal adaptor proteins to transmit signals to downstream cellular events, controlling the stress response, signal transduction, cell proliferation and apoptosis212, 213, 214, 215, 216. Previous investigations have demonstrated that HPK1 participates in practically every phase of the cancer immune cycle as a negative regulator. Inhibiting tumor growth is one of the effects of impaired HPK1 kinase activity, which also improves T cell signaling, virus clearance, and cytokine release. A recent study analyzed that HPK1 kinase activity inhibits immune function in a variety of cells, including CD4+ T cells, CD8+ T cells, and DCs, and confirmed that inactivation of its kinase domain is sufficient to induce an antitumor immune response217. Hernandez et al.217 identified the kinase-dependent effects of HPK1 in CD1 T cells by using HPK1 kinase-dead (HPK1.kd) knock-in mice. It has been demonstrated that the intrinsically increased T cell receptor signaling and cytokine release are caused by the loss of HPK1 kinase activity. In response to tumor challenges, HPK1.kd mice showed enhanced tumor growth inhibition and increased CD8 T cell function. Interestingly, the simultaneous blockade of PD-L1 and HPK1 was found to further enhance the function of effector T cells, thus providing better anti-tumor immunity than single-target blockade. These researches suggest that HPK1 is a promising target for cancer immunotherapy.

There is currently no drug on the market worldwide that targets HPK1, but several small molecule inhibitors have entered clinical trials.

7.1.1. CFI-402411-based combinational therapy

CFI-402411, an HPK1 inhibitor currently being investigated in phase II clinical trials for treating advanced solid tumors, has shown promising antitumor activities in mouse models of solid and hematologic malignancies when used as monotherapy or in combination with checkpoint inhibitors. For example, CFI-402411 is effective as a single agent and acts synergistically with anti-PD1 antibodies in CT26 tumor-bearing mice. Intriguingly, experimental tumor rechallenges after prior complete remission failed to result in tumor regrowth in mice treated with HPK1 inhibitors, indicating the immune memory induced by HPK1 inhibitors218. The safety and tolerability of CFI-402411 alone or in combination with pembrolizumab in patients with advanced solid tumors are being assessed in a first-in-human study (NCT04521413).

7.1.2. BGB-15025-based combinational therapy

BGB-15025 is another HPK1 inhibitor currently under phase I clinical investigation for advanced solid tumor (NCT04649385) (Fig. 14A), which aimed to examine the safety, pharmacokinetics, and preliminary anticancer efficacy of BGB-15025 alone and in combination with tislelizumab (anti-PD1 monoclonal antibody) in patients with advanced solid tumors.

Figure 14.

Figure 14

Representative inhibitors targeting immune-associated kinases, RORγt, and other targets. (A) Representative HPK1 inhibitor. (B) Representative RON inhibitor. (C) Representative RORγt inhibitors. (D) Representative inhibitors targeting other immunotherapy targets, including BET inhibitors (RO6870810, JQ1, and I-BET151), DHODH inhibitors (orludodstat and brequinar), and S1PR1 modulator (FTY720).

7.1.3. PF-07265028-based combinational therapy

For advanced or metastatic solid tumors, the HPK1 inhibitor PF-07265028 is presently being tested in a phase I clinical study (NCT05233436). This study's goal was to assess the effectiveness and safety of PF-07265028 monotherapy and in combination with sasanlimab (anti-PD-1 monoclonal antibody), establishing the maximum tolerated dosage (MTD) of PF-07265028. The suggested dose of PF-07265028 was chosen for monotherapy and combination therapy to assess the clinical efficacy of these treatments in future investigations.

7.1.4. PRJ1-3024-based combinational therapy

PRJ1-3024 is being tested in a phase II clinical trial for advanced solid tumors (NCT05315167). This study is designed to determine the MTD of PRJ1-3024 and assess its safety, tolerability, pharmacokinetics, and pharmacodynamics among patients with advanced solid tumors.

7.1.5. NDI-101150-based combinational therapy

Another HPK1 inhibitor, NDI-101150, is being studied in a phase I/II clinical trial (NCT05128487). The study was performed to evaluate the safety and initial antitumor activity of NDI-101150 as monotherapy or in combination with pembrolizumab in adult patients with advanced solid tumors. In addition, the MTD and recommended phase II dose were also identified.

7.1.6. GRC-54276-based combinational therapy

The HPK1 inhibitor, GRC-54276 is being investigated for advanced solid tumors and lymphomas in a phase I clinical study (NCT05878691). This research was conducted to evaluate the efficacy of GRC-54276 alone and in combination with pembrolizumab or atezolizumab (an anti-PD-L1 antibody) in patients with advanced solid tumors and lymphomas. Additionally, the MTD and suggested phase II doses of GRC-54276 alone and in combination with pembrolizumab or atezolizumab will also be determined.

7.1.7. RGT-264-based combinational therapy

RGT-264 is an HPK1 inhibitor being tested in a phase I clinical trial for advanced solid tumors (NCT05764915). This trial assesses the preliminary effectiveness, pharmacokinetics, safety, and tolerability of RGT-264 phosphate tablets in patients with advanced solid tumors. Besides, the MTD and the phase II dose advised for monotherapy were also evaluated.

7.2. Targeting RON signaling

Receptor tyrosine kinases (RTKs) have a significant impact on various cellular processes including cell growth, proliferation, differentiation, migration, metabolism, and cell cycle progression in cancer219,220. Recepteur d'origine nantais (RON, also known as macrophage stimulating 1 receptor, MST1R) expressed by epithelial-derived cells, osteoclasts, and macrophages, is an RTK of the Met proto-oncogene family which is a subfamily of RTKs containing another member, c-Mesenchymal–epithelial transition factor (c-Met)221. Macrophage-stimulating protein (MSP, also known as human macrophage stimulating 1, MST1, or mouse hepatocyte growth factor-like protein, HGFL) is currently recognized as the sole identified specific ligand of RON according to receptor binding, crosslinking, phosphorylation and induction of cell migration222.

The expression of RON was reported to be substantially higher in cancer cells than in normal epithelial cells. Crystal structure analysis revealed that one MSP molecule interacts with two RON semaphorins (SEMA)223. Interestingly, while ligand-dependent activation of RON occurs classically in normal tissues, RON can be activated by ligand-independent dimerization mainly in RON-overexpressing cancers222. Moreover, the activation of RON requires the presence of CD44, especially the v6-containing isoforms as coreceptors, which are also overexpressed in tumors224. Currently, RON overexpression has been observed in many primary tumors due to numerous mechanisms. RON transcription could be enhanced by additional transcription factors including NF-κB and HIF-1α in cancer cells225,226. Meanwhile, the methylation patterns in the RON promoter could be altered to favor transcription. Interestingly, RON overexpression is often accompanied by the generation of multiple RON isoforms. Among all discovered RON isoforms, RONΔ165, RONΔ160, and RONΔ155 were identified from primary human colorectal adenocarcinomas to drive cancer progression227, and the short-form Ron (sf RON) was shown to contribute to breast cancer pathogenesis228.

RON signaling activates downstream pathways such as P13K–AKT, RAS–ERK, and MAPK pathways, which subsequently stimulate distinct signaling cascades that are crucial for epithelial-to-mesenchymal transition, including TGFβ, STAT, β-catenin, and NF-κB pathways to facilitate carcinogenesis222. The remarkable crosstalk of active RON signaling with other oncogenic signaling pathways as a result of RON overexpression in tumors highlights its importance in tumorigenesis229. Furthermore, RON-activated signaling in stromal cells, especially TAM in the TME also contributes to tumor growth and invasiveness230. Recently, RON activation was shown to block antitumor CD8+ T cell response and thus contributed to immunotherapy resistance and metastasis231. Moreover, aberrant RON activation (via overexpression of RON protein or multiple RON isoforms) and MSP-RON signaling has been reported to induce and promote tumor survival, growth, metastasis, and chemoresistance in various cancers228,232, 233, 234, 235.

Given the significant role of RON signaling in cancer progression, targeting RON signaling has emerged as an attractive strategy for cancer therapy. Previous studies have demonstrated the antitumor effect by inhibition of RON signaling alone. Recently, accumulating evidence suggested that the combination of multi-tyrosine kinase inhibitors and RON inhibition may produce a more pronounced antitumor efficacy than targeting RON alone236. It is noteworthy that specific inhibitors of RON alone have not been discovered because RON shares similar protein structures with c-Met and various other RTKs237. Therefore, studies about small-molecule dual inhibitors of RON and c-Met were mainly reported in the literatures and they were mainly focused on the blockade of the HGF-c-Met signaling146,147. RON signaling was proposed as a co-therapeutic target due to its relatively weak kinase activities.

BMS-777607 (ASLAN002) is a selective inhibitor of RON kinase that has advanced into clinical trials because of its preclinical favorable pharmacokinetic and safety profiles and robust efficacy in vivo (Fig. 14B)238. The combination of BMS-777607 and anti-PD-1 was reported to significantly decrease tumor growth and incidence of lung metastasis vs. either monotherapy alone in a mouse model of triple-negative breast cancer239. Breast cancer cells are known to secrete high levels of MSP and activate the MSP-RON signaling to cause bone destruction. To this end, BMS-777607 was shown in a first-in-human clinical trial to reduce bone destruction and alter markers of bone turnover, which provided a rationale to target RON for breast cancer therapy240. However, Sharma et al.241 reported that BMS-777607 could increase resistance to cytotoxic chemotherapy agents by inducing polyploidy in breast cancer cell lines242. These studies pointed to the potential clinical application and complications when using RON kinase inhibitors for cancer treatment.

In a phase I multiple ascending dose study among patients with advanced or metastatic solid tumors, BMS-777607 was well tolerated and 300 mg twice daily was determined as the recommended dose for future phase II studies (RP2D)243. Another phase I/II trial (NCT00605618) investigating multiple ascending doses of BMS-777607 in patients with advanced or metastatic solid tumors is also ongoing.

8. Targeting RORγt

RORγt, a crucial transcription factor in the RORs (Retinoic acid receptor-related orphan receptors) family244, has been demonstrated to undergo sumoylation, which in turn facilitates the differentiation of naïve CD4+ T cells, leading to the generation of Th17 cells245. The differentiated Th17 cells could produce interleukin (IL)-17 and activate the anti-tumor response of CD8+ T cells (Fig. 15)246. CD8+ T cells are of utmost importance in tumor immunotherapy, as they fulfill crucial roles among all types of immune cells247. Unfortunately, the tumor microenvironment often employs various immune escape mechanisms that result in the suppression of the normal anti-tumor functions of CD8+ T cells248. Given that RORγt agonists have been demonstrated to activate RORγt signaling, facilitate the generation of Th17 cells, and promote the activation of CD8+ T cells, they have emerged as a novel strategy in immunotherapy (Fig. 14C)249. RORγt is a particularly attractive target among all IL-17 regulators (e.g., STAT3, IRF4, and BATF) because RORγt has a canonical NR ligand binding domain (LBD) that is readily accessible for the binding of small molecules250.

Figure 15.

Figure 15

The role of RORγt agonists in antitumor immunity. RORγt agonists have been demonstrated to upregulate the expression of CXCL10 in DCs, increase the population of CD8+ T cells, and activate NK cells. Moreover, it can promote the differentiation of CD4+ T cells into Th17 cells, which subsequently secrete IL-17 to activate CD8+ T cells. MDSCs, myeloid-derived suppressor cells; DCs, dendritic cells. Created with BioRender.com.

A recent study has revealed an alternative mechanism of RORγt agonists to potentiate immunotherapy249. The RORγt agonist (8-074) has been observed to upregulate CXCL10 expression in DCs and enhance the population of CD8+ T cells. In transwell experiments, elevated CXCL10 expression was shown to enhance the migration of CD8+ T cells. Crucially, the utilization of RORγt agonists has demonstrated the ability to significantly improve the effectiveness of anti-PD-1 immunotherapy. The enhancement effect of 8-074 was dependent on CXCL10 because the abundance and migration of CD8+ T cells were not affected if CXCL10 was neutralized in the tumor microenvironment.

The most commonly used RORγt agonists include steroids (e.g., cholesterol and desmosterol), synthetic aryl amide, and thiazole amide244. LYC-55716 is a first-in-class oral RORγt agonist currently in phase II clinical evaluation to test its efficacy against metastatic cancers (NCT02929862). Furthermore, the effectiveness of LYC-55716 as a monotherapy and in combination with pembrolizumab is also under active clinical investigation for the treatment of NSCLC (NCT03396497).

9. Small-molecule agents targeting other immunotherapy targets

9.1. Bromodomain inhibitors

The bromodomain is a protein domain that can recognize and bind to acetylated lysine residues present in various proteins important for cellular functions. Abnormal acetylation levels and dysfunction of the bromodomain-containing proteins could lead to dysregulation of gene transcription, which is associated with the development of various disorders including malignant tumors and inflammation. Among the protein families containing bromodomain, BET proteins, and CBP/p300 proteins are currently the most studied.

BET (Bromodomain and extra terminal) proteins, including BRD2, BRD3, BRD4, and BRDT, are involved in various biological processes and related to tumor development251. BET inhibitors exhibited potential antitumor effects, and some of them are under assessment in ongoing clinical trials. In the phase Ib study (NCT03255096), the combination of the BET inhibitor (RO6870810) and venetoclax with or without rituximab for the treatment of DLBCL showed an ORR of 38.5% and a complete response rate of 20.5%252. In another phase I clinical study of RO6870810 alone for the treatment of NUT carcinoma, other solid tumors, and DLBCL, the ORRs were 25% (2/8), 2% (1/47), and 11% (2/19), respectively253. Studies have also shown that BET bromodomain inhibitors JQ1 and I-BET151 can upregulate MICA to activate NK cells and regulate antitumor immunity254. Moreover, JQ1 was also reported to attenuate T regulatory cell suppressive function and synergize with the HDAC6 inhibitor ricolinostat to promote immune-mediated tumor growth arrest in NSCLC255. Furthermore, the combination of ricolinostat and JQ1 showed a significant inhibitory effect on SCLC tumor growth and this effect depended on NK cells256.

CBP (cAMP-responsive element-binding protein-binding protein) and p300 (E1A-associated protein p300) are highly homologous, so they are generally referred to as CBP/p300. CBP/p300 belongs to the histone acetyltransferase family. The bromodomain inhibitor CBP30 selectively inhibits CBP/p300 and reduces the secretion of cytokines such as IL-17A257. Other CBP/p300 bromodomain inhibitors CPI703 and CPI644 were also found to inhibit the differentiation of Treg cells and the cytokine secretion of Th17 cells, possibly related to the regulation of FOXP3 acetylation258. In addition, the bromodomain inhibitor GNE-781 can downregulate MYC and FOXP3, thereby inhibiting the function of Treg cells to enhance antitumor immunity259.

9.2. DHODH inhibitors

Dihydroorotate dehydrogenase (DHODH) is a key enzyme in the synthesis of pyrimidine nucleotides, which catalyzes the fourth step in the de novo pathway of pyrimidine nucleotide synthesis260. It is located on the inner mitochondrial membrane. Pyrimidine nucleotides can be synthesized by two pathways, the salvage pathway and the de novo pathway. In quiescent and fully differentiated cells, pyrimidine nucleotides are mainly synthesized by the salvage pathway. On the other hand, in rapidly proliferating cells such as malignant tumor cells and activated immune cells, the de novo pathway is activated as the demand for pyrimidine nucleotides increases to support cell proliferation. Therefore, the inhibition of DHODH may be exploited as a novel therapeutic strategy to inhibit abnormal cell proliferation.

DHODH inhibitors can inhibit the proliferation of activated lymphocytes by blocking the synthesis of pyrimidine nucleotides. Therefore, DHODH inhibitors are currently mainly used as immunosuppressants for the treatment of autoimmune diseases such as rheumatoid arthritis and multiple sclerosis261,262. The FDA has approved leflunomide and teriflunomide for clinical use.

In the field of cancer, DHODH inhibitors have also emerged as novel anticancer agents. In vivo and in vitro experiments showed that orludodstat (BAY2402234) can inhibit proliferation and induce differentiation in AML, which supported the initiation of phase I clinical trial (NCT03404726) to evaluate its safety, tolerability, maximum tolerated dose, and pharmacological active dose in AML, myelodysplastic syndrome, and chronic myeloid leukemia263. Brequinar has also been reported to induce differentiation of AML cells264. Recent preclinical studies suggested that DHODH inhibitors may also play a role in melanoma, glioblastoma, SCLC, neuroblastoma, and other tumors265, 266, 267, 268. In addition, the anticancer effect of DHODH inhibitors was shown to be related to the upregulation of p53 levels and ferroptosis269,270. Most recently, DHODH inhibitors were also reported to enhance the anti-tumor effect of ICI by inhibiting MDSCs271.

9.3. S1PR modulators

S1P (sphingosine-1-phosphate) receptors are a class of transmembrane G protein-coupled receptors, including five subtypes, that exert various effects by binding to their natural ligand S1P. It is an important regulator in inflammation, angiogenesis, vascular permeability, cancer growth, and metastasis. S1P receptor agonists are currently mainly used as immunosuppressive agents for autoimmune diseases, organ transplantation, etc.

FTY720 is a sphingosine analog, which has been clinically approved for the treatment of multiple sclerosis. It can activate all S1PRs except S1P2 after phosphorylation by sphingosine kinase272. Furthermore, FTY720 may act as a functional inhibitor of S1P1 by mediating the internalization of S1PRs, so its action may be bidirectional273. By targeting S1PRs, FTY720 can increase the retention of lymphocytes in lymph nodes and exert immunosuppressive effects274. In addition, FTY720 was also reported to exhibit antitumor activity, but these effects are likely independent of S1PRs and may involve other pathways such as SPHK5, PP2A6, etc.275, 276, 277. Representative small-molecule agents targeting these targets are summarized in Fig. 14D.

10. Conclusion and future perspective

ICIs, especially monoclonal antibodies that target PD-1/PD-L1 and CTLA-4, have significantly advanced cancer treatment and substantially improved patient outcomes. Unfortunately, only 30%–40% of patients benefit from ICIs, and resistance to ICI therapy is common. Accumulating evidence indicates that combination therapy with ICIs and other therapeutic means could significantly improve the therapeutic efficacy. Small molecule-based immunomodulators targeting various intracellular pathways have gained significant advances recently and can provide complementary or alternative therapies with ICIs or chemotherapy. An increasing number of small molecules that target PRR-associated pathways, immune checkpoint, oncogenic signaling, metabolic pathways, etc. have been developed, hoping to provide new treatment options for patients who progressed from prior immunotherapy as well as patients who appear unresponsive to ICI therapy. Multiple clinical studies are ongoing for small-molecule immunomodulators alone or in combination with ICIs in a variety of cancer types.

By comparison with mAbs, immunomodulatory small-molecule inhibitors have several advantages, such as better organ or tumor penetration, higher stability, and the ability to cross cell membranes. Although quite optimistic, dozens of clinical trials evaluating different types of immunomodulators have failed to provide solid evidence of patient benefit. The major drawback is the disappointing clinical efficacy, and this may be due to a lack of predictive biomarkers to better select appropriate patients. For instance, results from ECHO-301, the first large phase III trial to evaluate epacadostat combined with pembrolizumab for the treatment of advanced melanoma showed no benefit278. Possible causes including its impact on the TME are not well understood and lack of direct evidence for target inhibition. The anti-tumor efficacy of immunomodulatory small molecule inhibitors depends on cancer types and the tumor microenvironment. Specific molecular changes or mutations may affect therapeutic efficacy, and identifying specific genes/proteins and understanding the molecular mechanism of action will help to improve the efficacy of current immunomodulators.

Another drawback is adverse side effects caused by off-target toxicity or “cytokine storm”. Recently, the phase I trial of XMT-2056 (a HER2-targeted immunosynthen STING-agonist antibody–drug conjugate) has been suspended due to serious adverse events (NCT05514717). Both continuous activation of the cGAS–STING pathway and systemic administration of TLR agonists can lead to excessive production of cytokines, causing severe toxicity. In light of existing problems, the development of small-molecule immuno-oncology drugs is still in a premature state, further optimization is needed to increase efficiency, specificity, and safety, to expand its clinical application.

Although there are some disadvantages, the potential of immunomodulatory small molecule inhibitors did show great clinical benefit in several tumor types. Lenvatinib plus pembrolizumab significantly prolonged the survival of patients with advanced RCC (NCT02811861). In patients with unresectable HCC, treatment with the combination of camrelizumab and rivoceranib demonstrated a strong improvement in survival outcome (NCT03764293). These small molecules possess unique properties to increase the efficacy of cancer immunotherapy strategies while eliminating immunosuppression and tolerance. In this review, we summarized clinical trials of small-molecule immunomodulators alone or in combination with other treatments for cancer therapy. A synergistic antitumor effect could be achieved by some combinations. Currently, the FDA has approved 16 different immunomodulators including 9 checkpoint inhibitors (7 targeting the PD1/PD-L1 signaling pathway, 1 targeting the CD28/CTLA-4 pathway, and 1 targeting LAG-3), 4 cytokines (3 targeting the IFNAR1/2 pathway and 1 targeting the IL-2/IL-2R pathway), 2 immune adjuvants (1 targeting TLR7 and another targeting TLR3), and pexidartinib (a small molecule with immunomodulatory properties targeting KIT, CSF1R, and FLT3 pathways) for the treatment of different cancer types. Among them, it is encouraging to see that two TLR agonists (imiquimod and Poly ICLC), have been approved by the FDA as adjuvants for the clinical treatment of basal cell carcinoma and squamous cell carcinoma. Lenvatinib plus pembrolizumab has been approved for first-line treatment of adult patients with advanced RCC. The combination therapy of rivoceranib and camrelizumab was approved by the China NMPA as a first-line treatment for liver cancer and the application for FDA approval was reviewed.

In summary, with increasing research in TME and reliable biomarkers in guiding clinical immuno-oncology, more effective new molecules and combination strategies are expected.

Acknowledgements

This work was supported by the National Natural Science Foundation of China (Nos. U21A20421, 82073882, 82073317, 81772540 and 82272996), the Key Project of Science Technology Program of Guangzhou (No. 2023B03J0029, China), the National Key R&D Program of China (No. 2022YFE0209700), the Science and Technology Program of Guangzhou (Nos. 202201010819 and 202206010081, China).

Footnotes

Peer review under the responsibility of Chinese Pharmaceutical Association and Institute of Materia Medica, Chinese Academy of Medical Sciences.

Contributor Information

Kenneth Kin Wah To, Email: kennethto@cuhk.edu.hk.

Chenglai Xia, Email: xiachenglai@smu.edu.cn.

Jianye Zhang, Email: jianyez@gzhmu.edu.cn.

Zhi Shi, Email: tshizhi@jnu.edu.cn.

Zeping Hu, Email: zeping_hu@tsinghua.edu.cn.

Min Huang, Email: mhuang@simm.ac.cn.

Liwu Fu, Email: fulw@mail.sysu.edu.cn.

Author contributions

Liwu Fu conceived the study and revised the manuscript. Kenneth Kin Wah To, Jianye Zhang, Zhi Shi, Zeping Hu and Min Huang revised the manuscript. Fang Wang, Kai Fu, Yujue Wang, Can Pan, Zeyu Liu, Xiaopeng Li, Yu Lu and Chenglai Xia retrieved the related literatures and wrote the manuscript. Kai Fu, Can Pan and Yujue Wang created the pictures. Fang Wang, Kai Fu, Yujue Wang, Xueping Wang and Chuan Yang retrieved the clinical trials and produced the tables. All authors have read and approved this version of manuscript.

Conflicts of interest

The authors declare no conflicts of interest.

References

  • 1.Coley W.B. The diagnosis and treatment of bone sarcoma. Glasgow Med J. 1936;126:128–164. [PMC free article] [PubMed] [Google Scholar]
  • 2.O'Malley W.E., Achinstein B., Shear M.J. Action of bacterial polysaccharide on tumors. II. Damage of sarcoma 37 by serum of mice treated with Serratia marcescens polysaccharide, and induced tolerance. J Natl Cancer Inst. 1962;29 doi: 10.1111/j.1753-4887.1988.tb05376.x. Nutr Rev 1988;46:389-391. [DOI] [PubMed] [Google Scholar]
  • 3.Riley R.S., June C.H., Langer R., Mitchell M.J. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov. 2019;18:175–196. doi: 10.1038/s41573-018-0006-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Couzin-Frankel J. Breakthrough of the year 2013. Cancer immunotherapy. Science. 2013;342:1432–1433. doi: 10.1126/science.342.6165.1432. [DOI] [PubMed] [Google Scholar]
  • 5.Carlino M.S., Larkin J., Long G.V. Immune checkpoint inhibitors in melanoma. Lancet. 2021;398:1002–1014. doi: 10.1016/S0140-6736(21)01206-X. [DOI] [PubMed] [Google Scholar]
  • 6.Yi M., Zheng X., Niu M., Zhu S., Ge H., Wu K. Combination strategies with PD-1/PD-L1 blockade: current advances and future directions. Mol Cancer. 2022;21:28. doi: 10.1186/s12943-021-01489-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Haslam A., Prasad V. Estimation of the percentage of US patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. JAMA Netw Open. 2019;2 doi: 10.1001/jamanetworkopen.2019.2535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Yamaguchi H., Hsu J.M., Yang W.H., Hung M.C. Mechanisms regulating PD-L1 expression in cancers and associated opportunities for novel small-molecule therapeutics. Nat Rev Clin Oncol. 2022;19:287–305. doi: 10.1038/s41571-022-00601-9. [DOI] [PubMed] [Google Scholar]
  • 9.de Miguel M., Calvo E. Clinical challenges of immune checkpoint inhibitors. Cancer Cell. 2020;38:326–333. doi: 10.1016/j.ccell.2020.07.004. [DOI] [PubMed] [Google Scholar]
  • 10.Sun L., Wu J., Du F., Chen X., Chen Z.J. Cyclic GMP–AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science. 2013;339:786–791. doi: 10.1126/science.1232458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ding C., Song Z., Shen A., Chen T., Zhang A. Small molecules targeting the innate immune cGAS–STING–TBK1 signaling pathway. Acta Pharm Sin B. 2020;10:2272–2298. doi: 10.1016/j.apsb.2020.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Amouzegar A., Chelvanambi M., Filderman J.N., Storkus W.J., Luke J.J. STING agonists as cancer therapeutics. Cancers (Basel) 2021;13:2695. doi: 10.3390/cancers13112695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wu Y., Yang Z., Cheng K., Bi H., Chen J. Small molecule-based immunomodulators for cancer therapy. Acta Pharm Sin B. 2022;12:4287–4308. doi: 10.1016/j.apsb.2022.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Meric-Bernstam F., Sweis R.F., Hodi F.S., Messersmith W.A., Andtbacka R.H.I., Ingham M., et al. Phase I dose-escalation trial of MIW815 (ADU-S100), an intratumoral STING agonist, in patients with advanced/metastatic solid tumors or lymphomas. Clin Cancer Res. 2022;28:677–688. doi: 10.1158/1078-0432.CCR-21-1963. [DOI] [PubMed] [Google Scholar]
  • 15.Chang W., Altman M.D., Lesburg C.A., Perera S.A., Piesvaux J.A., Schroeder G.K., et al. Discovery of MK-1454: a potent cyclic dinucleotide stimulator of interferon genes agonist for the treatment of cancer. J Med Chem. 2022;65:5675–5689. doi: 10.1021/acs.jmedchem.1c02197. [DOI] [PubMed] [Google Scholar]
  • 16.Gremel G., mpagnatiello M.A., Carotta S., Schaaf O., Chetta P.M., Oost T., et al. Abstract 4522: potent induction of a tumor-specific immune response by a cyclic dinucleotide STING agonist. Am Assoc Cancer Res. 2020;80:4522. [Google Scholar]
  • 17.Challa S.V., Zhou S., Sheri A., Padmanabhan S., Meher G., Gimi R., et al. Preclinical studies of SB 11285, a novel STING agonist for immuno-oncology. J Clin Oncol. 2017;35 [Google Scholar]
  • 18.Cunniff E.C., Sato Y., Mai D., Appleman V.A., Iwasaki S., Kolev V., et al. TAK-676: a novel stimulator of interferon genes (STING) agonist promoting durable IFN-dependent antitumor immunity in preclinical studies. Cancer Res Commun. 2022;2:489–502. doi: 10.1158/2767-9764.CRC-21-0161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Pan B.S., Perera S.A., Piesvaux J.A., Presland J.P., Schroeder G.K., Cumming J.N., et al. An orally available non-nucleotide STING agonist with antitumor activity. Science. 2020;369 doi: 10.1126/science.aba6098. [DOI] [PubMed] [Google Scholar]
  • 20.Chin E.N., Yu C., Vartabedian V.F., Jia Y., Kumar M., Gamo A.M., et al. Antitumor activity of a systemic STING-activating non-nucleotide cGAMP mimetic. Science. 2020;369:993–999. doi: 10.1126/science.abb4255. [DOI] [PubMed] [Google Scholar]
  • 21.Ramanjulu J.M., Pesiridis G.S., Yang J., Concha N., Singhaus R., Zhang S.Y., et al. Design of amidobenzimidazole STING receptor agonists with systemic activity. Nature. 2018;564:439–443. doi: 10.1038/s41586-018-0705-y. [DOI] [PubMed] [Google Scholar]
  • 22.Kim D.S., Endo A., Fang F.G., Huang K.C., Bao X., Choi H.W., et al. E7766, a macrocycle-bridged stimulator of interferon genes (STING) agonist with potent pan-genotypic activity. ChemMedChem. 2021;16:1740–1743. doi: 10.1002/cmdc.202100068. [DOI] [PubMed] [Google Scholar]
  • 23.Allen B.K., Kulkarni M.M., Chamberlain B., Dwight T., Koh C., Samant R., et al. Design of a systemic small molecule clinical STING agonist using physics-based simulations and artificial intelligence. bioRxiv. 2022 doi: 10.1101/2022.05.23.493001. Available from: [DOI] [Google Scholar]
  • 24.Li D., Wu M. Pattern recognition receptors in health and diseases. Signal Transduct Target Ther. 2021;6:291. doi: 10.1038/s41392-021-00687-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Takeuchi O., Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–820. doi: 10.1016/j.cell.2010.01.022. [DOI] [PubMed] [Google Scholar]
  • 26.O'Neill L.A., Golenbock D., Bowie A.G. The history of Toll-like receptors-redefining innate immunity. Nat Rev Immunol. 2013;13:453–460. doi: 10.1038/nri3446. [DOI] [PubMed] [Google Scholar]
  • 27.Yang Y., Li H., Fotopoulou C., Cunnea P., Zhao X. Toll-like receptor-targeted anti-tumor therapies: advances and challenges. Front Immunol. 2022;13 doi: 10.3389/fimmu.2022.1049340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zheng R., Ma J. Immunotherapeutic implications of Toll-like receptors activation in tumor microenvironment. Pharmaceutics. 2022;14:2285. doi: 10.3390/pharmaceutics14112285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Platten M., Bunse L., Wick A., Bunse T., Le Cornet L., Harting I., et al. A vaccine targeting mutant IDH1 in newly diagnosed glioma. Nature. 2021;592:463–468. doi: 10.1038/s41586-021-03363-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Filaci G., Fenoglio D., Nole F., Zanardi E., Tomasello L., Aglietta M., et al. Telomerase-based GX301 cancer vaccine in patients with metastatic castration-resistant prostate cancer: a randomized phase II trial. Cancer Immunol Immunother. 2021;70:3679–3692. doi: 10.1007/s00262-021-03024-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Frega G., Wu Q., Le Naour J., Vacchelli E., Galluzzi L., Kroemer G., et al. Trial watch: experimental TLR7/TLR8 agonists for oncological indications. Oncoimmunology. 2020;9 doi: 10.1080/2162402X.2020.1796002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ribas A., Medina T., Kirkwood J.M., Zakharia Y., Gonzalez R., Davar D., et al. Overcoming PD-1 blockade resistance with CpG-A Toll-like receptor 9 agonist vidutolimod in patients with metastatic melanoma. Cancer Discov. 2021;11:2998–3007. doi: 10.1158/2159-8290.CD-21-0425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sullivan R.J. For Whom the Bell Tolls? A Toll-like receptor 9 agonist's journey from vaccine adjuvant to promising agent in anti-PD-1-resistant melanoma. Cancer Discov. 2021;11:2960. doi: 10.1158/2159-8290.CD-21-1226. [DOI] [PubMed] [Google Scholar]
  • 34.Haymaker C., Johnson D.H., Murthy R., Bentebibel S.E., Uemura M.I., Hudgens C.W., et al. Tilsotolimod with ipilimumab drives tumor responses in anti-PD-1 refractory melanoma. Cancer Discov. 2021;11:1996–2013. doi: 10.1158/2159-8290.CD-20-1546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Speetjens F.M., Welters M.J.P., Slingerland M., de Vos van Steenwijk P., Roozen I.C.F.M., Boekestijn S., et al. Phase I trial to determine safety and immunogenicity of amplivant, a synthetic toll-like receptor 2 ligand, conjugated to two HPV16 E6 synthetic long peptides. J Clin Oncol. 2021;39:2614. [Google Scholar]
  • 36.Bhatia S., Miller N.J., Lu H., Longino N.V., Ibrani D., Shinohara M.M., et al. Intratumoral G100, a TLR4 agonist, induces antitumor immune responses and tumor regression in patients with Merkel cell carcinoma. Clin Cancer Res. 2019;25:1185–1195. doi: 10.1158/1078-0432.CCR-18-0469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.van Puffelen J.H., Keating S.T., Oosterwijk E., van der Heijden A.G., Netea M.G., Joosten L.A.B., et al. Trained immunity as a molecular mechanism for BCG immunotherapy in bladder cancer. Nat Rev Urol. 2020;17:513–525. doi: 10.1038/s41585-020-0346-4. [DOI] [PubMed] [Google Scholar]
  • 38.Ji N., Mukherjee N., Reyes R.M., Gelfond J., Javors M., Meeks J.J., et al. Rapamycin enhances BCG-specific γδ T cells during intravesical BCG therapy for non-muscle invasive bladder cancer: a randomized, double-blind study. J Immunother Cancer. 2021;9 doi: 10.1136/jitc-2020-001941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Offringa R., Kotzner L., Huck B., Urbahns K. The expanding role for small molecules in immuno-oncology. Nat Rev Drug Discov. 2022;21:821–840. doi: 10.1038/s41573-022-00538-9. [DOI] [PubMed] [Google Scholar]
  • 40.Koblish H.K., Wu L., Wang L.S., Liu P.C.C., Wynn R., Rios-Doria J., et al. Characterization of INCB086550: a potent and novel small-molecule PD-L1 inhibitor. Cancer Discov. 2022;12:1482–1499. doi: 10.1158/2159-8290.CD-21-1156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wang Y., Liu X., Zou X., Wang S., Luo L., Liu Y., et al. Metabolism and interspecies variation of IMMH-010, a programmed cell death ligand 1 inhibitor prodrug. Pharmaceutics. 2021;13:598. doi: 10.3390/pharmaceutics13050598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lastwika K.J., Wilson W., Li Q.K., Norris J., Xu H., Ghazarian S.R., et al. Control of PD-L1 expression by oncogenic activation of the AKT–mTOR pathway in non-small cell lung cancer. Cancer Res. 2016;76:227–238. doi: 10.1158/0008-5472.CAN-14-3362. [DOI] [PubMed] [Google Scholar]
  • 43.Stutvoet T.S., Kol A., de Vries E.G., de Bruyn M., Fehrmann R.S., Terwisscha van Scheltinga A.G., et al. MAPK pathway activity plays a key role in PD-L1 expression of lung adenocarcinoma cells. J Pathol. 2019;249:52–64. doi: 10.1002/path.5280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ikeda S., Okamoto T., Okano S., Umemoto Y., Tagawa T., Morodomi Y., et al. PD-L1 is upregulated by simultaneous amplification of the PD-L1 and JAK2 genes in non-small cell lung cancer. J Thorac Oncol. 2016;11:62–71. doi: 10.1016/j.jtho.2015.09.010. [DOI] [PubMed] [Google Scholar]
  • 45.Emran A.A., Chatterjee A., Rodger E.J., Tiffen J.C., Gallagher S.J., Eccles M.R., et al. Targeting DNA methylation and EZH2 activity to overcome melanoma resistance to immunotherapy. Trends Immunol. 2019;40:328–344. doi: 10.1016/j.it.2019.02.004. [DOI] [PubMed] [Google Scholar]
  • 46.Huang K.C., Chiang S.F., Chen W.T., Chen T.W., Hu C.H., Yang P.C., et al. Decitabine augments chemotherapy-induced PD-L1 upregulation for PD-L1 blockade in colorectal cancer. Cancers (Basel) 2020;12:462. doi: 10.3390/cancers12020462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zhu T., Hu Z., Wang Z., Ding H., Li R., Sun J., et al. Epigenetically silenced PD-L1 confers drug resistance to anti-PD1 therapy in gastric cardia adenocarcinoma. Int Immunopharmacol. 2020;82 doi: 10.1016/j.intimp.2020.106245. [DOI] [PubMed] [Google Scholar]
  • 48.Woods D.M., Sodre A.L., Villagra A., Sarnaik A., Sotomayor E.M., Weber J. HDAC inhibition upregulates PD-1 ligands in melanoma and augments immunotherapy with PD-1 blockade. Cancer Immunol Res. 2015;3:1375–1385. doi: 10.1158/2326-6066.CIR-15-0077-T. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Shi Y., Fu Y., Zhang X., Zhao G., Yao Y., Guo Y., et al. Romidepsin (FK228) regulates the expression of the immune checkpoint ligand PD-L1 and suppresses cellular immune functions in colon cancer. Cancer Immunol Immunother. 2021;70:61–73. doi: 10.1007/s00262-020-02653-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Llopiz D., Ruiz M., Villanueva L., Iglesias T., Silva L., Egea J., et al. Enhanced anti-tumor efficacy of checkpoint inhibitors in combination with the histone deacetylase inhibitor Belinostat in a murine hepatocellular carcinoma model. Cancer Immunol Immunother. 2019;68:379–393. doi: 10.1007/s00262-018-2283-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ni L., Dong C. New checkpoints in cancer immunotherapy. Immunol Rev. 2017;276:52–65. doi: 10.1111/imr.12524. [DOI] [PubMed] [Google Scholar]
  • 52.Mehta N., Maddineni S., Mathews I., Andres Parra Sperberg R., Huang P.S., Cochran J.R. Structure and functional binding epitope of V-domain Ig suppressor of T cell activation. Cell Rep. 2019;28 doi: 10.1016/j.celrep.2019.07.073. 2509–16.e5. [DOI] [PubMed] [Google Scholar]
  • 53.ElTanbouly M.A., Zhao Y., Nowak E., Li J., Schaafsma E., Le Mercier I., et al. VISTA is a checkpoint regulator for naive T cell quiescence and peripheral tolerance. Science. 2020;367 doi: 10.1126/science.aay0524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Wang L., Rubinstein R., Lines J.L., Wasiuk A., Ahonen C., Guo Y., et al. VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J Exp Med. 2011;208:577–592. doi: 10.1084/jem.20100619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Li N., Xu W., Yuan Y., Ayithan N., Imai Y., Wu X., et al. Immune-checkpoint protein VISTA critically regulates the IL-23/IL-17 inflammatory axis. Sci Rep. 2017;7:1485. doi: 10.1038/s41598-017-01411-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Le Mercier I., Chen W., Lines J.L., Day M., Li J., Sergent P., et al. VISTA regulates the development of protective antitumor immunity. Cancer Res. 2014;74:1933–1944. doi: 10.1158/0008-5472.CAN-13-1506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.ElTanbouly M.A., Schaafsma E., Noelle R.J., Lines J.L. VISTA: coming of age as a multi-lineage immune checkpoint. Clin Exp Immunol. 2020;200:120–130. doi: 10.1111/cei.13415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Musielak B., Kocik J., Skalniak L., Magiera-Mularz K., Sala D., Czub M., et al. CA-170—a potent small-molecule PD-L1 inhibitor or not?. Molecules. 2019;24:2804. doi: 10.3390/molecules24152804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Sasikumar P.G., Sudarshan N.S., Adurthi S., Ramachandra R.K., Samiulla D.S., Lakshminarasimhan A., et al. PD-1 derived CA-170 is an oral immune checkpoint inhibitor that exhibits preclinical anti-tumor efficacy. Commun Biol. 2021;4:699. doi: 10.1038/s42003-021-02191-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Powderly J., Patel M.R., Lee J.J., Brody J., Meric-Bernstam F., Hamilton E., et al. CA-170, a first in class oral small molecule dual inhibitor of immune checkpoints PD-L1 and VISTA, demonstrates tumor growth inhibition in pre-clinical models and promotes T cell activation in Phase 1 study. Ann Oncol. 2017;28:v405–v406. [Google Scholar]
  • 61.Radhakrishnan V., Bakhshi S., Prabhash K., Deshmukh C., Nag S., Lakshmaiah K. Phase 2 trial of CA-170, a novel oral small molecule dual inhibitor of immune checkpoints VISTA and PD-1, in patients (pts) with advanced solid tumor and Hodgkin lymphoma. J Immunother Cancer. 2018;6:P714. [Google Scholar]
  • 62.Pan J., Chen Y., Zhang Q., Khatun A., Palen K., Xin G., et al. Inhibition of lung tumorigenesis by a small molecule CA170 targeting the immune checkpoint protein VISTA. Commun Biol. 2021;4:906. doi: 10.1038/s42003-021-02381-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Li M.O., Wan Y.Y., Sanjabi S., Robertson A.K., Flavell R.A. Transforming growth factor-beta regulation of immune responses. Annu Rev Immunol. 2006;24:99–146. doi: 10.1146/annurev.immunol.24.021605.090737. [DOI] [PubMed] [Google Scholar]
  • 64.Derynck R., Turley S.J., Akhurst R.J. TGFβ biology in cancer progression and immunotherapy. Nat Rev Clin Oncol. 2021;18:9–34. doi: 10.1038/s41571-020-0403-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Tauriello D.V.F., Sancho E., Batlle E. Overcoming TGFβ-mediated immune evasion in cancer. Nat Rev Cancer. 2022;22:25–44. doi: 10.1038/s41568-021-00413-6. [DOI] [PubMed] [Google Scholar]
  • 66.Yingling J.M., Shou J., Xia X., Chen Y., Yan L., Evans W., et al. A small molecule inhibitor of TGFβ RI kinase potentiates VEGF dependent angiogenesis in vitro. Cancer Res. 2006;66:59. [Google Scholar]
  • 67.Santini V., Valcarcel D., Platzbecker U., Komrokji R.S., Cleverly A.L., Lahn M.M., et al. Phase II study of the ALK5 inhibitor galunisertib in very low-, low-, and intermediate-risk myelodysplastic syndromes. Clin Cancer Res. 2019;25:6976–6985. doi: 10.1158/1078-0432.CCR-19-1338. [DOI] [PubMed] [Google Scholar]
  • 68.Harding J.J., Do R.K., Yaqubie A., Cleverly A., Zhao Y., Gueorguieva I., et al. Phase 1b study of galunisertib and ramucirumab in patients with advanced hepatocellular carcinoma. Cancer Med. 2021;10:3059–3067. doi: 10.1002/cam4.3880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wick A., Desjardins A., Suarez C., Forsyth P., Gueorguieva I., Burkholder T., et al. Phase 1b/2a study of galunisertib, a small molecule inhibitor of transforming growth factor-beta receptor I, in combination with standard temozolomide-based radiochemotherapy in patients with newly diagnosed malignant glioma. Invest New Drugs. 2020;38:1570–1579. doi: 10.1007/s10637-020-00910-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Melisi D., Oh D.Y., Hollebecque A., Calvo E., Varghese A., Borazanci E., et al. Safety and activity of the TGFβ receptor I kinase inhibitor galunisertib plus the anti-PD-L1 antibody durvalumab in metastatic pancreatic cancer. J Immunother Cancer. 2021;9 doi: 10.1136/jitc-2020-002068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Yamazaki T., Gunderson A.J., Gilchrist M., Whiteford M., Kiely M.X., Hayman A., et al. Galunisertib plus neoadjuvant chemoradiotherapy in patients with locally advanced rectal cancer: a single-arm, phase 2 trial. Lancet Oncol. 2022;23:1189–1200. doi: 10.1016/S1470-2045(22)00446-6. [DOI] [PubMed] [Google Scholar]
  • 72.Nadal E., Saleh M., Aix S.P., Ochoa-de-Olza M., Patel S.P., Antonia S., et al. A phase Ib/II study of galunisertib in combination with nivolumab in solid tumors and non-small cell lung cancer. BMC Cancer. 2023;23:708. doi: 10.1186/s12885-023-11153-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Brandes A.A., Carpentier A.F., Kesari S., Sepulveda-Sanchez J.M., Wheeler H.R., Chinot O., et al. A phase II randomized study of galunisertib monotherapy or galunisertib plus lomustine compared with lomustine monotherapy in patients with recurrent glioblastoma. Neuro Oncol. 2016;18:1146–1156. doi: 10.1093/neuonc/now009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Xu G., Zhang Y., Wang H., Guo Z., Wang X., Li X., et al. Synthesis and biological evaluation of 4-(pyridin-4-oxy)-3-(3,3-difluorocyclobutyl)-pyrazole derivatives as novel potent transforming growth factor-β type 1 receptor inhibitors. Eur J Med Chem. 2020;198 doi: 10.1016/j.ejmech.2020.112354. [DOI] [PubMed] [Google Scholar]
  • 75.Yap T.A., Vieito M., Baldini C., Sepulveda-Sanchez J.M., Kondo S., Simonelli M., et al. First-in-human phase I study of a next-generation, oral, TGFβ receptor 1 inhibitor, LY3200882, in patients with advanced cancer. Clin Cancer Res. 2021;27:6666–6676. doi: 10.1158/1078-0432.CCR-21-1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Hong E., Barczak W., Park S., Heo J.S., Ooshima A., Munro S., et al. Combination treatment of T1-44, a PRMT5 inhibitor with vactosertib, an inhibitor of TGF-β signaling, inhibits invasion and prolongs survival in a mouse model of pancreatic tumors. Cell Death Dis. 2023;14:93. doi: 10.1038/s41419-023-05630-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Son J.Y., Park S.Y., Kim S.J., Lee S.J., Park S.A., Kim M.J., et al. EW-7197, a novel ALK-5 kinase inhibitor, potently inhibits breast to lung metastasis. Mol Cancer Ther. 2014;13:1704–1716. doi: 10.1158/1535-7163.MCT-13-0903. [DOI] [PubMed] [Google Scholar]
  • 78.Yoon J.H., Jung S.M., Park S.H., Kato M., Yamashita T., Lee I.K., et al. Activin receptor-like kinase5 inhibition suppresses mouse melanoma by ubiquitin degradation of Smad4, thereby derepressing eomesodermin in cytotoxic T lymphocytes. EMBO Mol Med. 2013;5:1720–1739. doi: 10.1002/emmm.201302524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Park S., Yang K.M., Park Y., Hong E., Hong C.P., Park J., et al. Identification of epithelial–mesenchymal transition-related target genes induced by the mutation of smad3 linker phosphorylation. J Cancer Prev. 2018;23:1–9. doi: 10.15430/JCP.2018.23.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kim T.W., Lee K.W., Ahn J.B., Park Y.S., Ock C.Y., Park H., et al. 74 Tumor microenvironment based on PD-L1 and CD8 T-cell infiltration correlated with the response of MSS mCRC patients treated vactosertib in combination with pembrolizumab. J Immunother Cancer. 2021;9:A82. [Google Scholar]
  • 81.Choi J., Park J., Cho I., Sheen Y. Co-treatment with vactosertib, a novel, orally bioavailable activin receptor-like kinase 5 inhibitor, suppresses radiotherapy-induced epithelial-to-mesenchymal transition, cancer cell stemness, and lung metastasis of breast cancer. Radiol Oncol. 2022;56:185–197. doi: 10.2478/raon-2022-0012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Hu G., Zhao R., Zhou F., Lan J., Zheng B., Lu Q., et al. Abstract 3072: GFH018, a novel TGF-βRI inhibitor, for the treatment of advanced solid tumors. Cancer Res. 2019;79 [Google Scholar]
  • 83.Tremblay D., Mascarenhas J. Next generation therapeutics for the treatment of myelofibrosis. Cells. 2021;10 doi: 10.3390/cells10051034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Hume D.A., MacDonald K.P. Therapeutic applications of macrophage colony-stimulating factor-1 (CSF-1) and antagonists of CSF-1 receptor (CSF-1R) signaling. Blood. 2012;119:1810–1820. doi: 10.1182/blood-2011-09-379214. [DOI] [PubMed] [Google Scholar]
  • 85.Han J., Chitu V., Stanley E.R., Wszolek Z.K., Karrenbauer V.D., Harris R.A. Inhibition of colony stimulating factor-1 receptor (CSF-1R) as a potential therapeutic strategy for neurodegenerative diseases: opportunities and challenges. Cell Mol Life Sci. 2022;79:219. doi: 10.1007/s00018-022-04225-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Xiang C., Li H., Tang W. Targeting CSF-1R represents an effective strategy in modulating inflammatory diseases. Pharmacol Res. 2023;187 doi: 10.1016/j.phrs.2022.106566. [DOI] [PubMed] [Google Scholar]
  • 87.Lamb Y.N. Pexidartinib: first approval. Drugs. 2019;79:1805–1812. doi: 10.1007/s40265-019-01210-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Tap W.D., Gelderblom H., Palmerini E., Desai J., Bauer S., Blay J.Y., et al. Pexidartinib versus placebo for advanced tenosynovial giant cell tumour (ENLIVEN): a randomised phase 3 trial. Lancet. 2019;394:478–487. doi: 10.1016/S0140-6736(19)30764-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Butowski N., Colman H., De Groot J.F., Omuro A.M., Nayak L., Wen P.Y., et al. Orally administered colony stimulating factor 1 receptor inhibitor PLX3397 in recurrent glioblastoma: an ivy foundation early phase clinical trials consortium phase II study. Neuro Oncol. 2016;18:557–564. doi: 10.1093/neuonc/nov245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Manji G.A., Van Tine B.A., Lee S.M., Raufi A.G., Pellicciotta I., Hirbe A.C., et al. A phase I study of the combination of pexidartinib and sirolimus to target tumor-associated macrophages in unresectable sarcoma and malignant peripheral nerve sheath tumors. Clin Cancer Res. 2021;27:5519–5527. doi: 10.1158/1078-0432.CCR-21-1779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Xu J., Shen L., Zhou Z., Li J., Bai C., Chi Y., et al. Surufatinib in advanced extrapancreatic neuroendocrine tumours (SANET-ep): a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 2020;21:1500–1512. doi: 10.1016/S1470-2045(20)30496-4. [DOI] [PubMed] [Google Scholar]
  • 92.Xu J., Shen L., Bai C., Wang W., Li J., Yu X., et al. Surufatinib in advanced pancreatic neuroendocrine tumours (SANET-p): a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 2020;21:1489–1499. doi: 10.1016/S1470-2045(20)30493-9. [DOI] [PubMed] [Google Scholar]
  • 93.Sun Y., Yang L., Hao X., Liu Y., Zhang J., Ning Z., et al. Phase I dose-escalation study of chiauranib, a novel angiogenic, mitotic, and chronic inflammation inhibitor, in patients with advanced solid tumors. J Hematol Oncol. 2019;12:9. doi: 10.1186/s13045-018-0695-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Johnson M., Dudek A.Z., Sukari A., Call J., Kunk P.R., Lewis K., et al. ARRY-382 in combination with pembrolizumab in patients with advanced solid tumors: results from a phase 1b/2 study. Clin Cancer Res. 2022;28:2517–2526. doi: 10.1158/1078-0432.CCR-21-3009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.von Tresckow B., Morschhauser F., Ribrag V., Topp M.S., Chien C., Seetharam S., et al. An open-label, multicenter, phase I/II study of JNJ-40346527, a CSF-1R inhibitor, in patients with relapsed or refractory hodgkin lymphoma. Clin Cancer Res. 2015;21:1843–1850. doi: 10.1158/1078-0432.CCR-14-1845. [DOI] [PubMed] [Google Scholar]
  • 96.Propper D.J., Balkwill F.R. Harnessing cytokines and chemokines for cancer therapy. Nat Rev Clin Oncol. 2022;19:237–253. doi: 10.1038/s41571-021-00588-9. [DOI] [PubMed] [Google Scholar]
  • 97.Karin N. Chemokines and cancer: new immune checkpoints for cancer therapy. Curr Opin Immunol. 2018;51:140–145. doi: 10.1016/j.coi.2018.03.004. [DOI] [PubMed] [Google Scholar]
  • 98.Nywening T.M., Wang-Gillam A., Sanford D.E., Belt B.A., Panni R.Z., Cusworth B.M., et al. Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol. 2016;17:651–662. doi: 10.1016/S1470-2045(16)00078-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Noel M., O'Reilly E.M., Wolpin B.M., Ryan D.P., Bullock A.J., Britten C.D., et al. Phase 1b study of a small molecule antagonist of human chemokine (C–C motif) receptor 2 (PF-04136309) in combination with nab-paclitaxel/gemcitabine in first-line treatment of metastatic pancreatic ductal adenocarcinoma. Invest New Drugs. 2020;38:800–811. doi: 10.1007/s10637-019-00830-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Linehan D., Noel M.S., Hezel A.F., Wang-Gillam A., Eskens F., Sleijfer S., et al. Overall survival in a trial of orally administered CCR2 inhibitor CCX872 in locally advanced/metastatic pancreatic cancer: correlation with blood monocyte counts. J Clin Oncol. 2018;36:92. [Google Scholar]
  • 101.Le D., Gutierrez M.E., Saleh M., Chen E., Mallick A.B., Pishvaian M.J., et al. Abstract CT124: a phase Ib/II study of BMS-813160, a CC chemokine receptor (CCR) 2/5 dual antagonist, in combination with chemotherapy or nivolumab in patients (pts) with advanced pancreatic or colorectal cancer. Cancer Res. 2018;78:CT124. [Google Scholar]
  • 102.Berlato C., Khan M.N., Schioppa T., Thompson R., Maniati E., Montfort A., et al. A CCR4 antagonist reverses the tumor-promoting microenvironment of renal cancer. J Clin Invest. 2017;127:801–813. doi: 10.1172/JCI82976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Aldinucci D., Borghese C., Casagrande N. The CCL5/CCR5 axis in cancer progression. Cancers (Basel) 2020;12 doi: 10.3390/cancers12071765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Haag G.M., Halama N., Springfeld C., Grün B., Apostolidis L., Zschaebitz S., et al. Combined PD-1 inhibition (Pembrolizumab) and CCR5 inhibition (Maraviroc) for the treatment of refractory microsatellite stable (MSS) metastatic colorectal cancer (mCRC): first results of the PICCASSO phase I trial. J Clin Oncol. 2020;38 [Google Scholar]
  • 105.Haag G.M., Springfeld C., Grun B., Apostolidis L., Zschabitz S., Dietrich M., et al. Pembrolizumab and maraviroc in refractory mismatch repair proficient/microsatellite-stable metastatic colorectal cancer—the PICCASSO phase I trial. Eur J Cancer. 2022;167:112–122. doi: 10.1016/j.ejca.2022.03.017. [DOI] [PubMed] [Google Scholar]
  • 106.Bilusic M., Heery C.R., Collins J.M., Donahue R.N., Palena C., Madan R.A., et al. Phase I trial of HuMax-IL8 (BMS-986253), an anti-IL-8 monoclonal antibody, in patients with metastatic or unresectable solid tumors. J Immunother Cancer. 2019;7:240. doi: 10.1186/s40425-019-0706-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Davar D., Simonelli M., Gutierrez M., Calvo E., Melear J., Piha-Paul S., et al. 394 Interleukin-8–neutralizing monoclonal antibody BMS-986253 plus nivolumab (NIVO) in biomarker-enriched, primarily anti-PD-(L)1-experienced patients with advanced cancer: initial phase 1 results. J Immunother Cancer. 2020;8:A239–A240. [Google Scholar]
  • 108.Shi Y., Riese D.J., 2nd, Shen J. The role of the CXCL12/CXCR4/CXCR7 chemokine axis in cancer. Front Pharmacol. 2020;11 doi: 10.3389/fphar.2020.574667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Zhou Y., Cao H.B., Li W.J., Zhao L. The CXCL12 (SDF-1)/CXCR4 chemokine axis: oncogenic properties, molecular targeting, and synthetic and natural product CXCR4 inhibitors for cancer therapy. Chin J Nat Med. 2018;16:801–810. doi: 10.1016/S1875-5364(18)30122-5. [DOI] [PubMed] [Google Scholar]
  • 110.Pernas S., Martin M., Kaufman P.A., Gil-Martin M., Gomez Pardo P., Lopez-Tarruella S., et al. Balixafortide plus eribulin in HER2-negative metastatic breast cancer: a phase 1, single-arm, dose-escalation trial. Lancet Oncol. 2018;19:812–824. doi: 10.1016/S1470-2045(18)30147-5. [DOI] [PubMed] [Google Scholar]
  • 111.Hainsworth J.D., Reeves J.A., Mace J.R., Crane E.J., Hamid O., Stille J.R., et al. A randomized, open-label phase 2 study of the CXCR4 inhibitor LY2510924 in combination with sunitinib versus sunitinib alone in patients with metastatic renal cell carcinoma (RCC) Target Oncol. 2016;11:643–653. doi: 10.1007/s11523-016-0434-9. [DOI] [PubMed] [Google Scholar]
  • 112.O'Hara M.H., Messersmith W., Kindler H., Zhang W., Pitou C., Szpurka A.M., et al. Safety and pharmacokinetics of CXCR4 peptide antagonist, LY2510924, in combination with durvalumab in advanced refractory solid tumors. J Pancreat Cancer. 2020;6:21–31. doi: 10.1089/pancan.2019.0018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Bockorny B., Semenisty V., Macarulla T., Borazanci E., Wolpin B.M., Stemmer S.M., et al. BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: the COMBAT trial. Nat Med. 2020;26:878–885. doi: 10.1038/s41591-020-0880-x. [DOI] [PubMed] [Google Scholar]
  • 114.Caldwell R.W., Rodriguez P.C., Toque H.A., Narayanan S.P., Caldwell R.B. Arginase: a multifaceted enzyme important in health and disease. Physiol Rev. 2018;98:641–665. doi: 10.1152/physrev.00037.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Shyer J.A., Flavell R.A., Bailis W. Metabolic signaling in T cells. Cell Res. 2020;30:649–659. doi: 10.1038/s41422-020-0379-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Vonwirth V., Bulbul Y., Werner A., Echchannaoui H., Windschmitt J., Habermeier A., et al. Inhibition of arginase 1 liberates potent T cell immunostimulatory activity of human neutrophil granulocytes. Front Immunol. 2021;11 doi: 10.3389/fimmu.2020.617699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Albaugh V.L., Pinzon-Guzman C., Barbul A. Arginine-dual roles as an onconutrient and immunonutrient. J Surg Oncol. 2017;115:273–280. doi: 10.1002/jso.24490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Ren W., Li Y., Xia X., Guo W., Zhai T., Jin Y., et al. Arginine inhibits the malignant transformation induced by interferon-gamma through the NF-kappaB-GCN2/eIF2alpha signaling pathway in mammary epithelial cells in vitro and in vivo. Exp Cell Res. 2018;368:236–247. doi: 10.1016/j.yexcr.2018.05.003. [DOI] [PubMed] [Google Scholar]
  • 119.Hong Y., Wen R., Wu G., Li S., Liu W., Chen Z., et al. Abnormal immune function of MDSC and NK cells from chronic phase CML patients restores with tyrosine kinase inhibitors. Int Immunopharmacol. 2022;109 doi: 10.1016/j.intimp.2022.108821. [DOI] [PubMed] [Google Scholar]
  • 120.Zhang J., Xu X., Shi M., Chen Y., Yu D., Zhao C., et al. CD13hi neutrophil-like myeloid-derived suppressor cells exert immune suppression through Arginase 1 expression in pancreatic ductal adenocarcinoma. Oncoimmunology. 2017;6 doi: 10.1080/2162402X.2016.1258504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Czystowska-Kuzmicz M., Sosnowska A., Nowis D., Ramji K., Szajnik M., Chlebowska-Tuz J., et al. Small extracellular vesicles containing arginase-1 suppress T-cell responses and promote tumor growth in ovarian carcinoma. Nat Commun. 2019;10:3000. doi: 10.1038/s41467-019-10979-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Grzybowski M.M., Pęczkowicz-Szyszka J., Wolska P., Stańczak P.S., Welzer M., Nikolaev E., et al. Targeting ARG2 as a novel therapeutic approach for cancer. Ann Oncol. 2019;30:v793–v794. [Google Scholar]
  • 123.Marti i Lindez A.A., Dunand-Sauthier I., Conti M., Gobet F., Nunez N., Hannich J.T., et al. Mitochondrial arginase-2 is a cell-autonomous regulator of CD8+ T cell function and antitumor efficacy. JCI Insight. 2019;4 doi: 10.1172/jci.insight.132975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Dunand-Sauthier I., Irla M., Carnesecchi S., Seguin-Estevez Q., Vejnar C.E., Zdobnov E.M., et al. Repression of arginase-2 expression in dendritic cells by microRNA-155 is critical for promoting T cell proliferation. J Immunol. 2014;193:1690–1700. doi: 10.4049/jimmunol.1301913. [DOI] [PubMed] [Google Scholar]
  • 125.Zhai L., Ladomersky E., Lenzen A., Nguyen B., Patel R., Lauing K.L., et al. IDO1 in cancer: a Gemini of immune checkpoints. Cell Mol Immunol. 2018;15:447–457. doi: 10.1038/cmi.2017.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Liu M., Wang X., Wang L., Ma X., Gong Z., Zhang S., et al. Targeting the IDO1 pathway in cancer: from bench to bedside. J Hematol Oncol. 2018;11:100. doi: 10.1186/s13045-018-0644-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Amobi-McCloud A., Muthuswamy R., Battaglia S., Yu H., Liu T., Wang J., et al. IDO1 expression in ovarian cancer induces PD-1 in T cells via aryl hydrocarbon receptor activation. Front Immunol. 2021;12 doi: 10.3389/fimmu.2021.678999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Zhai L., Ladomersky E., Dostal C.R., Lauing K.L., Swoap K., Billingham L.K., et al. Non-tumor cell IDO1 predominantly contributes to enzyme activity and response to CTLA-4/PD-L1 inhibition in mouse glioblastoma. Brain Behav Immun. 2017;62:24–29. doi: 10.1016/j.bbi.2017.01.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Zhai L., Ladomersky E., Lauing K.L., Wu M., Genet M., Gritsina G., et al. Infiltrating T cells increase IDO1 expression in glioblastoma and contribute to decreased patient survival. Clin Cancer Res. 2017;23:6650–6660. doi: 10.1158/1078-0432.CCR-17-0120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Mitchell T.C., Hamid O., Smith D.C., Bauer T.M., Wasser J.S., Olszanski A.J., et al. Epacadostat plus pembrolizumab in patients with advanced solid tumors: phase I results from a multicenter, open-label phase I/II trial (ECHO-202/KEYNOTE-037) J Clin Oncol. 2018;36:3223–3230. doi: 10.1200/JCO.2018.78.9602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Blair A.B., Kleponis J., Thomas D.L., 2nd, Muth S.T., Murphy A.G., Kim V., et al. IDO1 inhibition potentiates vaccine-induced immunity against pancreatic adenocarcinoma. J Clin Invest. 2019;129:1742–1755. doi: 10.1172/JCI124077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Zakharia Y., McWilliams R.R., Rixe O., Drabick J., Shaheen M.F., Grossmann K.F., et al. Phase II trial of the IDO pathway inhibitor indoximod plus pembrolizumab for the treatment of patients with advanced melanoma. J Immunother Cancer. 2021;9 doi: 10.1136/jitc-2020-002057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Chen C., Guan J., Gu X., Chu Q., Zhu H. Prostaglandin E2 and receptors: insight into tumorigenesis, tumor progression, and treatment of hepatocellular carcinoma. Front Cell Dev Biol. 2022;10 doi: 10.3389/fcell.2022.834859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Zelenay S., van der Veen A.G., Böttcher J.P., Snelgrove K.J., Rogers N., Acton S.E., et al. Cyclooxygenase-dependent tumor growth through evasion of immunity. Cell. 2015;162:1257–1270. doi: 10.1016/j.cell.2015.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Kabir T.D., Leigh R.J., Tasena H., Mellone M., Coletta R.D., Parkinson E.K., et al. A miR-335/COX-2/PTEN axis regulates the secretory phenotype of senescent cancer-associated fibroblasts. Aging (Albany NY) 2016;8:1608–1635. doi: 10.18632/aging.100987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Tjiu J.W., Chen J.S., Shun C.T., Lin S.J., Liao Y.H., Chu C.Y., et al. Tumor-associated macrophage-induced invasion and angiogenesis of human basal cell carcinoma cells by cyclooxygenase-2 induction. J Invest Dermatol. 2009;129:1016–1025. doi: 10.1038/jid.2008.310. [DOI] [PubMed] [Google Scholar]
  • 137.Rong X., Huang B., Qiu S., Li X., He L., Peng Y. Tumor-associated macrophages induce vasculogenic mimicry of glioblastoma multiforme through cyclooxygenase-2 activation. Oncotarget. 2016;7:83976–83986. doi: 10.18632/oncotarget.6930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Gan L., Qiu Z., Huang J., Li Y., Huang H., Xiang T., et al. Cyclooxygenase-2 in tumor-associated macrophages promotes metastatic potential of breast cancer cells through Akt pathway. Int J Biol Sci. 2016;12:1533–1543. doi: 10.7150/ijbs.15943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Hashemi Goradel N., Najafi M., Salehi E., Farhood B., Mortezaee K. Cyclooxygenase-2 in cancer: a review. J Cell Physiol. 2019;234:5683–5699. doi: 10.1002/jcp.27411. [DOI] [PubMed] [Google Scholar]
  • 140.Obermajer N., Muthuswamy R., Lesnock J., Edwards R.P., Kalinski P. Positive feedback between PGE2 and COX2 redirects the differentiation of human dendritic cells toward stable myeloid-derived suppressor cells. Blood. 2011;118:5498–5505. doi: 10.1182/blood-2011-07-365825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Böttcher J.P., Bonavita E., Chakravarty P., Blees H., Cabeza-Cabrerizo M., Sammicheli S., et al. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell. 2018;172 doi: 10.1016/j.cell.2018.01.004. 1022–37.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Bonavita E., Bromley C.P., Jonsson G., Pelly V.S., Sahoo S., Walwyn-Brown K., et al. Antagonistic inflammatory phenotypes dictate tumor fate and response to immune checkpoint blockade. Immunity. 2020;53 doi: 10.1016/j.immuni.2020.10.020. 1215–29.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Meyerhardt J.A., Shi Q., Fuchs C.S., Meyer J., Niedzwiecki D., Zemla T., et al. Effect of celecoxib vs placebo added to standard adjuvant therapy on disease-free survival among patients with stage III colon cancer: the CALGB/SWOG 80702 (Alliance) randomized clinical trial. JAMA. 2021;325:1277–1286. doi: 10.1001/jama.2021.2454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Coombes R.C., Tovey H., Kilburn L., Mansi J., Palmieri C., Bartlett J., et al. Effect of celecoxib vs placebo as adjuvant therapy on disease-free survival among patients with breast cancer: the REACT randomized clinical trial. JAMA Oncol. 2021;7:1291–1301. doi: 10.1001/jamaoncol.2021.2193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Hu H., Kang L., Zhang J., Wu Z., Wang H., Huang M., et al. Neoadjuvant PD-1 blockade with toripalimab, with or without celecoxib, in mismatch repair-deficient or microsatellite instability-high, locally advanced, colorectal cancer (PICC): a single-centre, parallel-group, non-comparative, randomised, phase 2 trial. Lancet Gastroenterol Hepatol. 2022;7:38–48. doi: 10.1016/S2468-1253(21)00348-4. [DOI] [PubMed] [Google Scholar]
  • 146.Huang L., Jiang S., Shi Y. Tyrosine kinase inhibitors for solid tumors in the past 20 years (2001–2020) J Hematol Oncol. 2020;13:143. doi: 10.1186/s13045-020-00977-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Mh E.-W., Hm A., Mn S., Am H., Im L. Design, synthesis and molecular modeling studies of new series of antitumor 1,2,4-triazines with potential c-Met kinase inhibitory activity. Bioorg Chem. 2018;76:154–165. doi: 10.1016/j.bioorg.2017.11.006. [DOI] [PubMed] [Google Scholar]
  • 148.Churov A., Zhulai G. Targeting adenosine and regulatory T cells in cancer immunotherapy. Hum Immunol. 2021;82:270–278. doi: 10.1016/j.humimm.2020.12.005. [DOI] [PubMed] [Google Scholar]
  • 149.Kayhan M., Koyas A., Akdemir I., Savas A.C., Cekic C. Adenosine receptor signaling targets both PKA and Epac pathways to polarize dendritic cells to a suppressive phenotype. J Immunol. 2019;203:3247–3255. doi: 10.4049/jimmunol.1900765. [DOI] [PubMed] [Google Scholar]
  • 150.Wei Q., Zhang L., Zhao N., Cheng Z., Xin H., Ding J. Immunosuppressive adenosine-targeted biomaterials for emerging cancer immunotherapy. Front Immunol. 2022;13 doi: 10.3389/fimmu.2022.1012927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Congreve M., Brown G.A., Borodovsky A., Lamb M.L. Targeting adenosine A2A receptor antagonism for treatment of cancer. Expert Opin Drug Discov. 2018;13:997–1003. doi: 10.1080/17460441.2018.1534825. [DOI] [PubMed] [Google Scholar]
  • 152.Borodovsky A., Barbon C.M., Wang Y., Ye M., Prickett L., Chandra D., et al. Small molecule AZD4635 inhibitor of A2AR signaling rescues immune cell function including CD103+ dendritic cells enhancing anti-tumor immunity. J Immunother Cancer. 2020;8 doi: 10.1136/jitc-2019-000417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Sitkovsky M.V. Lessons from the A2a adenosine receptor antagonist-enabled tumor regression and survival in patients with treatment-refractory renal cell cancer. Cancer Discov. 2020;10:16–19. doi: 10.1158/2159-8290.CD-19-1280. [DOI] [PubMed] [Google Scholar]
  • 154.Lim E.A., Bendell J.C., Falchook G.S., Bauer T.M., Drake C.G., Choe J.H., et al. Phase Ia/b, open-label, multicenter study of AZD4635 (an adenosine A2a receptor antagonist) as monotherapy or combined with durvalumab, in patients with solid tumors. Clin Cancer Res. 2022;28:4871–4884. doi: 10.1158/1078-0432.CCR-22-0612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Seitz L., Jin L., Leleti M., Ashok D., Jeffrey J., Rieger A., et al. Safety, tolerability, and pharmacology of AB928, a novel dual adenosine receptor antagonist, in a randomized, phase 1 study in healthy volunteers. Invest New Drugs. 2019;37:711–721. doi: 10.1007/s10637-018-0706-6. [DOI] [PubMed] [Google Scholar]
  • 156.Subudhi S.K., Bendell J.C., Carducci M.A., Kopp L.M., Scott J., Grady M.M., et al. ARC-6: a phase 1b/2, open-label, randomized platform study to evaluate efficacy and safety of etrumadenant (AB928)-based treatment combinations in patients with metastatic castrate-resistant prostate cancer (mCRPC) J Clin Oncol. 2021;39 [Google Scholar]
  • 157.Cecchini M., Quah C., Liu S., Woloski R., Udyavar A., Giannakis M. Abstract CT129: ARC-3: updated results of etrumadenant (AB928) + modified FOLFOX-6 (mFOLFOX-6) in metastatic colorectal cancer (mCRC) patients. Cancer Res. 2021;81 [Google Scholar]
  • 158.Chiappori A.A., Creelan B., Tanvetyanon T., Gray J.E., Haura E.B., Thapa R., et al. Phase I study of taminadenant (PBF509/NIR178), an adenosine 2A receptor antagonist, with or without spartalizumab (PDR001), in patients with advanced non-small cell lung cancer. Clin Cancer Res. 2022;28:2313–2320. doi: 10.1158/1078-0432.CCR-21-2742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Okkenhaug K., Vanhaesebroeck B. PI3K in lymphocyte development, differentiation and activation. Nat Rev Immunol. 2003;3:317–330. doi: 10.1038/nri1056. [DOI] [PubMed] [Google Scholar]
  • 160.Lucas C.L., Chandra A., Nejentsev S., Condliffe A.M., Okkenhaug K. PI3Kδ and primary immunodeficiencies. Nat Rev Immunol. 2016;16:702–714. doi: 10.1038/nri.2016.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Kaneda M.M., Messer K.S., Ralainirina N., Li H., Leem C.J., Gorjestani S., et al. PI3Kγ is a molecular switch that controls immune suppression. Nature. 2016;539:437–442. doi: 10.1038/nature19834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Biswas S.K. Metabolic reprogramming of immune cells in cancer progression. Immunity. 2015;43:435–449. doi: 10.1016/j.immuni.2015.09.001. [DOI] [PubMed] [Google Scholar]
  • 163.Hao Y., Samuels Y., Li Q., Krokowski D., Guan B.J., Wang C., et al. Oncogenic PIK3CA mutations reprogram glutamine metabolism in colorectal cancer. Nat Commun. 2016;7 doi: 10.1038/ncomms11971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Hutti J.E., Pfefferle A.D., Russell S.C., Sircar M., Perou C.M., Baldwin A.S. Oncogenic PI3K mutations lead to NF-κB-dependent cytokine expression following growth factor deprivation. Cancer Res. 2012;72:3260–3269. doi: 10.1158/0008-5472.CAN-11-4141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Sun P., Meng L.H. Emerging roles of class I PI3K inhibitors in modulating tumor microenvironment and immunity. Acta Pharmacol Sin. 2020;41:1395–1402. doi: 10.1038/s41401-020-00500-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Vanhaesebroeck B., Perry M.W.D., Brown J.R., Andre F., Okkenhaug K. PI3K inhibitors are finally coming of age. Nat Rev Drug Discov. 2021;20:741–769. doi: 10.1038/s41573-021-00209-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Bosch A., Li Z., Bergamaschi A., Ellis H., Toska E., Prat A., et al. PI3K inhibition results in enhanced estrogen receptor function and dependence in hormone receptor-positive breast cancer. Sci Transl Med. 2015;7:283ra51. doi: 10.1126/scitranslmed.aaa4442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Andre F., Ciruelos E., Rubovszky G., Campone M., Loibl S., Rugo H.S., et al. Alpelisib for PIK3CA-mutated, hormone receptor-positive advanced breast cancer. N Engl J Med. 2019;380:1929–1940. doi: 10.1056/NEJMoa1813904. [DOI] [PubMed] [Google Scholar]
  • 169.Andre F., Ciruelos E.M., Juric D., Loibl S., Campone M., Mayer I.A., et al. Alpelisib plus fulvestrant for PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: final overall survival results from SOLAR-1. Ann Oncol. 2021;32:208–217. doi: 10.1016/j.annonc.2020.11.011. [DOI] [PubMed] [Google Scholar]
  • 170.Hanan E.J., Braun M.G., Heald R.A., MacLeod C., Chan C., Clausen S., et al. Discovery of GDC-0077 (inavolisib), a highly selective inhibitor and degrader of mutant PI3Kα. J Med Chem. 2022;65:16589–16621. doi: 10.1021/acs.jmedchem.2c01422. [DOI] [PubMed] [Google Scholar]
  • 171.Juric D., de Bono J.S., LoRusso P.M., Nemunaitis J., Heath E.I., Kwak E.L., et al. A first-in-human, phase I, dose-escalation study of TAK-117, a selective PI3Kα isoform inhibitor, in patients with advanced solid malignancies. Clin Cancer Res. 2017;23:5015–5023. doi: 10.1158/1078-0432.CCR-16-2888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Choueiri T.K., Porta C., Suárez C., Hainsworth J., Voog E., Duran I., et al. Randomized phase II trial of sapanisertib ± TAK-117 vs. everolimus in patients with advanced renal cell carcinoma after VEGF-targeted therapy. Oncologist. 2022;27:1048–1057. doi: 10.1093/oncolo/oyac192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Markham A. Idelalisib: first global approval. Drugs. 2014;74:1701–1707. doi: 10.1007/s40265-014-0285-6. [DOI] [PubMed] [Google Scholar]
  • 174.Fruman D.A., Cantley L.C. Idelalisib—a PI3Kδ inhibitor for B-cell cancers. N Engl J Med. 2014;370:1061–1062. doi: 10.1056/NEJMe1400055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Sharman J.P., Coutre S.E., Furman R.R., Cheson B.D., Pagel J.M., Hillmen P., et al. Final results of a randomized, phase III study of rituximab with or without idelalisib followed by open-label idelalisib in patients with relapsed chronic lymphocytic leukemia. J Clin Oncol. 2019;37:1391–1402. doi: 10.1200/JCO.18.01460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Zelenetz A.D., Barrientos J.C., Brown J.R., Coiffier B., Delgado J., Egyed M., et al. Idelalisib or placebo in combination with bendamustine and rituximab in patients with relapsed or refractory chronic lymphocytic leukaemia: interim results from a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2017;18:297–311. doi: 10.1016/S1470-2045(16)30671-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Jones J.A., Robak T., Brown J.R., Awan F.T., Badoux X., Coutre S., et al. Efficacy and safety of idelalisib in combination with ofatumumab for previously treated chronic lymphocytic leukaemia: an open-label, randomised phase 3 trial. Lancet Haematol. 2017;4:e114–e126. doi: 10.1016/S2352-3026(17)30019-4. [DOI] [PubMed] [Google Scholar]
  • 178.Lunning M., Vose J., Nastoupil L., Fowler N., Burger J.A., Wierda W.G., et al. Ublituximab and umbralisib in relapsed/refractory B-cell non-Hodgkin lymphoma and chronic lymphocytic leukemia. Blood. 2019;134:1811–1820. doi: 10.1182/blood.2019002118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Nastoupil L.J., Lunning M.A., Vose J.M., Schreeder M.T., Siddiqi T., Flowers C.R., et al. Tolerability and activity of ublituximab, umbralisib, and ibrutinib in patients with chronic lymphocytic leukaemia and non-Hodgkin lymphoma: a phase 1 dose escalation and expansion trial. Lancet Haematol. 2019;6:e100–e109. doi: 10.1016/S2352-3026(18)30216-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Forero-Torres A., Ramchandren R., Yacoub A., Wertheim M.S., Edenfield W.J., Caimi P., et al. Parsaclisib, a potent and highly selective PI3Kδ inhibitor, in patients with relapsed or refractory B-cell malignancies. Blood. 2019;133:1742–1752. doi: 10.1182/blood-2018-08-867499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Jiang B., Qi J., Song Y., Li Z., Tu M., Ping L., et al. Phase 1 clinical trial of the PI3Kδ inhibitor YY-20394 in patients with B-cell hematological malignancies. J Hematol Oncol. 2021;14:130. doi: 10.1186/s13045-021-01140-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Wang T., Sun X., Qiu L., Su H., Cao J., Li Z., et al. The oral PI3Kδ inhibitor linperlisib for the treatment of relapsed and/or refractory follicular lymphoma: a phase II, single-arm, open-label clinical trial. Clin Cancer Res. 2023;29:1440–1449. doi: 10.1158/1078-0432.CCR-22-2939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Jiang M., He K., Qiu T., Sun J., Liu Q., Zhang X., et al. Tumor-targeted delivery of silibinin and IPI-549 synergistically inhibit breast cancer by remodeling the microenvironment. Int J Pharm. 2020;581 doi: 10.1016/j.ijpharm.2020.119239. [DOI] [PubMed] [Google Scholar]
  • 184.De Henau O., Rausch M., Winkler D., Campesato L.F., Liu C., Cymerman D.H., et al. Overcoming resistance to checkpoint blockade therapy by targeting PI3Kγ in myeloid cells. Nature. 2016;539:443–447. doi: 10.1038/nature20554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Funk C.R., Wang S., Chen K.Z., Waller A., Sharma A., Edgar C.L., et al. PI3Kδ/γ inhibition promotes human CART cell epigenetic and metabolic reprogramming to enhance antitumor cytotoxicity. Blood. 2022;139:523–537. doi: 10.1182/blood.2021011597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Yoon Y.N., Lee E., Kwon Y.J., Gim J.A., Kim T.J., Kim J.S. PI3Kδ/γ inhibitor BR101801 extrinsically potentiates effector CD8+ T cell-dependent antitumor immunity and abscopal effect after local irradiation. J Immunother Cancer. 2022;10 doi: 10.1136/jitc-2021-003762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Dreyling M., Morschhauser F., Bouabdallah K., Bron D., Cunningham D., Assouline S.E., et al. Phase II study of copanlisib, a PI3K inhibitor, in relapsed or refractory, indolent or aggressive lymphoma. Ann Oncol. 2017;28:2169–2178. doi: 10.1093/annonc/mdx289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Dreyling M., Santoro A., Mollica L., Leppa S., Follows G.A., Lenz G., et al. Phosphatidylinositol 3-kinase inhibition by copanlisib in relapsed or refractory indolent lymphoma. J Clin Oncol. 2017;35:3898–3905. doi: 10.1200/JCO.2017.75.4648. [DOI] [PubMed] [Google Scholar]
  • 189.Yhim H.Y., Kim T., Kim S.J., Shin H.J., Koh Y., Kim J.S., et al. Combination treatment of copanlisib and gemcitabine in relapsed/refractory PTCL (COSMOS): an open-label phase I/II trial. Ann Oncol. 2021;32:552–559. doi: 10.1016/j.annonc.2020.12.009. [DOI] [PubMed] [Google Scholar]
  • 190.Matasar M.J., Capra M., Ozcan M., Lv F., Li W., Yanez E., et al. Copanlisib plus rituximab versus placebo plus rituximab in patients with relapsed indolent non-Hodgkin lymphoma (CHRONOS-3): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2021;22:678–689. doi: 10.1016/S1470-2045(21)00145-5. [DOI] [PubMed] [Google Scholar]
  • 191.Dent S., Cortes J., Im Y.H., Dieras V., Harbeck N., Krop I.E., et al. Phase III randomized study of taselisib or placebo with fulvestrant in estrogen receptor-positive, PIK3CA-mutant, HER2-negative, advanced breast cancer: the SANDPIPER trial. Ann Oncol. 2021;32:197–207. doi: 10.1016/j.annonc.2020.10.596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Flinn I.W., Hillmen P., Montillo M., Nagy Z., Illes A., Etienne G., et al. The phase 3 DUO trial: duvelisib vs ofatumumab in relapsed and refractory CLL/SLL. Blood. 2018;132:2446–2455. doi: 10.1182/blood-2018-05-850461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Locatelli S.L., Careddu G., Serio S., Consonni F.M., Maeda A., Viswanadha S., et al. Targeting cancer cells and tumor microenvironment in preclinical and clinical models of Hodgkin lymphoma using the dual PI3Kδ/γ inhibitor RP6530. Clin Cancer Res. 2019;25:1098–1112. doi: 10.1158/1078-0432.CCR-18-1133. [DOI] [PubMed] [Google Scholar]
  • 194.Soulieres D., Faivre S., Mesia R., Remenar E., Li S.H., Karpenko A., et al. Buparlisib and paclitaxel in patients with platinum-pretreated recurrent or metastatic squamous cell carcinoma of the head and neck (BERIL-1): a randomised, double-blind, placebo-controlled phase 2 trial. Lancet Oncol. 2017;18:323–335. doi: 10.1016/S1470-2045(17)30064-5. [DOI] [PubMed] [Google Scholar]
  • 195.Martin M., Chan A., Dirix L., O'Shaughnessy J., Hegg R., Manikhas A., et al. A randomized adaptive phase II/III study of buparlisib, a pan-class I PI3K inhibitor, combined with paclitaxel for the treatment of HER2- advanced breast cancer (BELLE-4) Ann Oncol. 2017;28:313–320. doi: 10.1093/annonc/mdw562. [DOI] [PubMed] [Google Scholar]
  • 196.Villarino A.V., Kanno Y., O'Shea J.J. Mechanisms and consequences of JAK–STAT signaling in the immune system. Nat Immunol. 2017;18:374–384. doi: 10.1038/ni.3691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Castelo-Soccio L., Kim H., Gadina M., Schwartzberg P.L., Laurence A., O'Shea J.J. Protein kinases: drug targets for immunological disorders. Nat Rev Immunol. 2023;23:787–806. doi: 10.1038/s41577-023-00877-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Johnson D.E., O'Keefe R.A., Grandis J.R. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol. 2018;15:234–248. doi: 10.1038/nrclinonc.2018.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.McLornan D.P., Pope J.E., Gotlib J., Harrison C.N. Current and future status of JAK inhibitors. Lancet. 2021;398:803–816. doi: 10.1016/S0140-6736(21)00438-4. [DOI] [PubMed] [Google Scholar]
  • 200.Hu X., Li J., Fu M., Zhao X., Wang W. The JAK/STAT signaling pathway: from bench to clinic. Signal Transduct Target Ther. 2021;6:402. doi: 10.1038/s41392-021-00791-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Harrison C., Kiladjian J.J., Al-Ali H.K., Gisslinger H., Waltzman R., Stalbovskaya V., et al. JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N Engl J Med. 2012;366:787–798. doi: 10.1056/NEJMoa1110556. [DOI] [PubMed] [Google Scholar]
  • 202.Pardanani A., Harrison C., Cortes J.E., Cervantes F., Mesa R.A., Milligan D., et al. Safety and efficacy of fedratinib in patients with primary or secondary myelofibrosis: a randomized clinical trial. JAMA Oncol. 2015;1:643–651. doi: 10.1001/jamaoncol.2015.1590. [DOI] [PubMed] [Google Scholar]
  • 203.Mesa R.A., Vannucchi A.M., Mead A., Egyed M., Szoke A., Suvorov A., et al. Pacritinib versus best available therapy for the treatment of myelofibrosis irrespective of baseline cytopenias (PERSIST-1): an international, randomised, phase 3 trial. Lancet Haematol. 2017;4:e225–e236. doi: 10.1016/S2352-3026(17)30027-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Verstovsek S., Mesa R.A., Gotlib J., Levy R.S., Gupta V., DiPersio J.F., et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366:799–807. doi: 10.1056/NEJMoa1110557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Harrison C.N., Garcia J.S., Somervaille T.C.P., Foran J.M., Verstovsek S., Jamieson C., et al. Addition of navitoclax to ongoing ruxolitinib therapy for patients with myelofibrosis with progression or suboptimal response: phase II safety and efficacy. J Clin Oncol. 2022;40:1671–1680. doi: 10.1200/JCO.21.02188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Harrison C.N., Schaap N., Vannucchi A.M., Kiladjian J.J., Tiu R.V., Zachee P., et al. Janus kinase-2 inhibitor fedratinib in patients with myelofibrosis previously treated with ruxolitinib (JAKARTA-2): a single-arm, open-label, non-randomised, phase 2, multicentre study. Lancet Haematol. 2017;4:e317–e324. doi: 10.1016/S2352-3026(17)30088-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Mascarenhas J., Hoffman R., Talpaz M., Gerds A.T., Stein B., Gupta V., et al. Pacritinib vs best available therapy, including ruxolitinib, in patients with myelofibrosis: a randomized clinical trial. JAMA Oncol. 2018;4:652–659. doi: 10.1001/jamaoncol.2017.5818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Harrison C.N., Vannucchi A.M., Platzbecker U., Cervantes F., Gupta V., Lavie D., et al. Momelotinib versus best available therapy in patients with myelofibrosis previously treated with ruxolitinib (SIMPLIFY 2): a randomised, open-label, phase 3 trial. Lancet Haematol. 2018;5:e73–e81. doi: 10.1016/S2352-3026(17)30237-5. [DOI] [PubMed] [Google Scholar]
  • 209.Mesa R.A., Kiladjian J.J., Catalano J.V., Devos T., Egyed M., Hellmann A., et al. SIMPLIFY-1: a phase III randomized trial of momelotinib versus ruxolitinib in Janus kinase inhibitor-naive patients with myelofibrosis. J Clin Oncol. 2017;35:3844–3850. doi: 10.1200/JCO.2017.73.4418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Verstovsek S., Gerds A.T., Vannucchi A.M., Al-Ali H.K., Lavie D., Kuykendall A.T., et al. Momelotinib versus danazol in symptomatic patients with anaemia and myelofibrosis (MOMENTUM): results from an international, double-blind, randomised, controlled, phase 3 study. Lancet. 2023;401:269–280. doi: 10.1016/S0140-6736(22)02036-0. [DOI] [PubMed] [Google Scholar]
  • 211.Lacey B.M., Xu Z., Chai X., Laskey J., Fradera X., Mittal P., et al. Development of high-throughput assays for evaluation of hematopoietic progenitor kinase 1 inhibitors. SLAS Discov. 2021;26:88–99. doi: 10.1177/2472555220952071. [DOI] [PubMed] [Google Scholar]
  • 212.Zhou L., Wang T., Zhang K., Zhang X., Jiang S. The development of small-molecule inhibitors targeting HPK1. Eur J Med Chem. 2022;244 doi: 10.1016/j.ejmech.2022.114819. [DOI] [PubMed] [Google Scholar]
  • 213.Wang Y., Zhang K., Georgiev P., Wells S., Xu H., Lacey B.M., et al. Pharmacological inhibition of hematopoietic progenitor kinase 1 positively regulates T-cell function. PLoS One. 2020;15 doi: 10.1371/journal.pone.0243145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Shui J.W., Boomer J.S., Han J., Xu J., Dement G.A., Zhou G., et al. Hematopoietic progenitor kinase 1 negatively regulates T cell receptor signaling and T cell-mediated immune responses. Nat Immunol. 2007;8:84–91. doi: 10.1038/ni1416. [DOI] [PubMed] [Google Scholar]
  • 215.Si J., Shi X., Sun S., Zou B., Li Y., An D., et al. Hematopoietic progenitor kinase1 (HPK1) mediates T cell dysfunction and is a druggable target for T cell-based immunotherapies. Cancer Cell. 2020;38 doi: 10.1016/j.ccell.2020.08.001. 551–66.e11. [DOI] [PubMed] [Google Scholar]
  • 216.Boomer J.S., Tan T.H. Functional interactions of HPK1 with adaptor proteins. J Cell Biochem. 2005;95:34–44. doi: 10.1002/jcb.20401. [DOI] [PubMed] [Google Scholar]
  • 217.Hernandez S., Qing J., Thibodeau R.H., Du X., Park S., Lee H.M., et al. The kinase activity of hematopoietic progenitor kinase 1 is essential for the regulation of T cell function. Cell Rep. 2018;25:80–94. doi: 10.1016/j.celrep.2018.09.012. [DOI] [PubMed] [Google Scholar]
  • 218.Linney I.D., Kaila N. Inhibitors of immuno-oncology target HPK1—a patent review (2016 to 2020) Expert Opin Ther Pat. 2021;31:893–910. doi: 10.1080/13543776.2021.1924671. [DOI] [PubMed] [Google Scholar]
  • 219.Du Z., Lovly C.M. Mechanisms of receptor tyrosine kinase activation in cancer. Mol Cancer. 2018;17:58. doi: 10.1186/s12943-018-0782-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Butti R., Das S., Gunasekaran V.P., Yadav A.S., Kumar D., Kundu G.C. Receptor tyrosine kinases (RTKs) in breast cancer: signaling, therapeutic implications and challenges. Mol Cancer. 2018;17 doi: 10.1186/s12943-018-0797-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Huang L., Fang X., Shi D., Yao S., Wu W., Fang Q., et al. MSP–RON pathway: potential regulator of inflammation and innate immunity. Front Immunol. 2020;11 doi: 10.3389/fimmu.2020.569082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Yao H.P., Zhou Y.Q., Zhang R., Wang M.H. MSP–RON signalling in cancer: pathogenesis and therapeutic potential. Nat Rev Cancer. 2013;13:466–481. doi: 10.1038/nrc3545. [DOI] [PubMed] [Google Scholar]
  • 223.Chao K.L., Tsai I.W., Chen C., Herzberg O. Crystal structure of the Sema-PSI extracellular domain of human RON receptor tyrosine kinase. PLoS One. 2012;7 doi: 10.1371/journal.pone.0041912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Matzke A., Herrlich P., Ponta H., Orian-Rousseau V. A five-amino-acid peptide blocks Met- and Ron-dependent cell migration. Cancer Res. 2005;65:6105–6110. doi: 10.1158/0008-5472.CAN-05-0207. [DOI] [PubMed] [Google Scholar]
  • 225.Thangasamy A., Rogge J., Ammanamanchi S. Recepteur d'origine nantais tyrosine kinase is a direct target of hypoxia-inducible factor-1alpha-mediated invasion of breast carcinoma cells. J Biol Chem. 2009;284:14001–14010. doi: 10.1074/jbc.M809320200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Zhao S., Ammanamanchi S., Brattain M., Cao L., Thangasamy A., Wang J., et al. Smad4-dependent TGF-beta signaling suppresses RON receptor tyrosine kinase-dependent motility and invasion of pancreatic cancer cells. J Biol Chem. 2008;283:11293–11301. doi: 10.1074/jbc.M800154200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Zhou Y.Q., He C., Chen Y.Q., Wang D., Wang M.H. Altered expression of the RON receptor tyrosine kinase in primary human colorectal adenocarcinomas: generation of different splicing RON variants and their oncogenic potential. Oncogene. 2003;22:186–197. doi: 10.1038/sj.onc.1206075. [DOI] [PubMed] [Google Scholar]
  • 228.Liu X., Zhao L., Derose Y.S., Lin Y.-C., Bieniasz M., Eyob H., et al. Short-form Ron promotes spontaneous breast cancer metastasis through interaction with phosphoinositide 3-kinase. Genes Cancer. 2011;2:753–762. doi: 10.1177/1947601911421924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Wagh P.K., Gray J.K., Zinser G.M., Vasiliauskas J., James L., Monga S.P., et al. β-Catenin is required for Ron receptor-induced mammary tumorigenesis. Oncogene. 2011;30:3694–3704. doi: 10.1038/onc.2011.86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Gurusamy D., Gray J.K., Pathrose P., Kulkarni R.M., Finkleman F.D., Waltz S.E. Myeloid-specific expression of Ron receptor kinase promotes prostate tumor growth. Cancer Res. 2013;73:1752–1763. doi: 10.1158/0008-5472.CAN-12-2474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Eyob H., Ekiz H.A., Welm A.L. RON promotes the metastatic spread of breast carcinomas by subverting antitumor immune responses. OncoImmunology. 2013;2 doi: 10.4161/onci.25670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Dustin D., Gu G., Beyer A.R., Herzog S.K., Edwards D.G., Lin H., et al. RON signalling promotes therapeutic resistance in ESR1 mutant breast cancer. Br J Cancer. 2021;124:191–206. doi: 10.1038/s41416-020-01174-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.McClaine R.J., Marshall A.M., Wagh P.K., Waltz S.E. Ron receptor tyrosine kinase activation confers resistance to tamoxifen in breast cancer cell lines. Neoplasia. 2010;12:650–658. doi: 10.1593/neo.10476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Bieniasz M., Radhakrishnan P., Faham N., De La O.J., Welm A.L. Preclinical efficacy of Ron kinase inhibitors alone and in combination with PI3K inhibitors for treatment of sfRon-expressing breast cancer patient-derived xenografts. Clin Cancer Res. 2015;21:5588–5600. doi: 10.1158/1078-0432.CCR-14-3283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Peace B.E., Toney-Earley K., Collins M.H., Waltz S.E. Ron receptor signaling augments mammary tumor formation and metastasis in a murine model of breast cancer. Cancer Res. 2005;65:1285–1293. doi: 10.1158/0008-5472.CAN-03-3580. [DOI] [PubMed] [Google Scholar]
  • 236.Baird A.-M., Easty D., Jarzabek M., Shiels L., Soltermann A., Klebe S., et al. When RON met TAM in mesothelioma: all druggable for one, and one drug for all? Front Endocrinol (Lausanne) 2019;10:89. doi: 10.3389/fendo.2019.00089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Zhang Y., Kaplan-Lefko P.J., Rex K., Yang Y., Moriguchi J., Osgood T., et al. Identification of a novel recepteur d’origine nantais/c-met small-molecule kinase inhibitor with antitumor activity in vivo. Cancer Res. 2008;68:6680–6687. doi: 10.1158/0008-5472.CAN-07-6782. [DOI] [PubMed] [Google Scholar]
  • 238.Schroeder G.M., An Y., Cai Z.-W., Chen X.-T., Clark C., Cornelius L.A.M., et al. Discovery of N-(4-(2-amino-3-chloropyridin-4-yloxy)-3-fluorophenyl)-4-ethoxy-1-(4-fluorophenyl)-2-oxo-1,2-dihydropyridine-3-carboxamide (BMS-777607), a selective and orally efficacious inhibitor of the Met kinase superfamily. J Med Chem. 2009;52:1251–1254. doi: 10.1021/jm801586s. [DOI] [PubMed] [Google Scholar]
  • 239.Kasikara C., Davra V., Calianese D., Geng K., Spires T.E., Quigley M., et al. Pan-TAM tyrosine kinase inhibitor BMS-777607 enhances anti-PD-1 mAb efficacy in a murine model of triple-negative breast cancer. Cancer Res. 2019;79:2669–2683. doi: 10.1158/0008-5472.CAN-18-2614. [DOI] [PubMed] [Google Scholar]
  • 240.Andrade K., Fornetti J., Zhao L., Miller S.C., Randall R.L., Anderson N., et al. RON kinase: a target for treatment of cancer-induced bone destruction and osteoporosis. Sci Transl Med. 2017;9 doi: 10.1126/scitranslmed.aai9338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Sharma S., Zeng J.Y., Zhuang C.M., Zhou Y.Q., Yao H.P., Hu X., et al. Small-molecule inhibitor BMS-777607 induces breast cancer cell polyploidy with increased resistance to cytotoxic chemotherapy agents. Mol Cancer Ther. 2013;12:725–736. doi: 10.1158/1535-7163.MCT-12-1079. [DOI] [PubMed] [Google Scholar]
  • 242.Sharma S., Yao H.P., Zhou Y.Q., Zhou J., Zhang R., Wang M.H. Prevention of BMS-777607-induced polyploidy/senescence by mTOR inhibitor AZD8055 sensitizes breast cancer cells to cytotoxic chemotherapeutics. Mol Oncol. 2014;8:469–482. doi: 10.1016/j.molonc.2013.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Roohullah A., Cooper A., Lomax A.J., Aung J., Barge A., Chow L., et al. A phase I trial to determine safety and pharmacokinetics of ASLAN002, an oral MET superfamily kinase inhibitor, in patients with advanced or metastatic solid cancers. Invest New Drugs. 2018;36:886–894. doi: 10.1007/s10637-018-0588-7. [DOI] [PubMed] [Google Scholar]
  • 244.Fan J., Lv Z., Yang G., Liao T.T., Xu J., Wu F., et al. Retinoic acid receptor-related orphan receptors: critical roles in tumorigenesis. Front Immunol. 2018;9 doi: 10.3389/fimmu.2018.01187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.He Z., Zhang J., Huang Z., Du Q., Li N., Zhang Q., et al. Sumoylation of RORγt regulates TH17 differentiation and thymocyte development. Nat Commun. 2018;9 doi: 10.1038/s41467-018-07203-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Zhong X., Wu H., Zhang W., Gwack Y., Shang W., Lee K.O., et al. Decoupling the role of RORγt in the differentiation and effector function of TH17 cells. Sci Adv. 2022;8 doi: 10.1126/sciadv.adc9221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Tsukumo S.I., Yasutomo K. Regulation of CD8+ T cells and antitumor immunity by Notch signaling. Front Immunol. 2018;9 doi: 10.3389/fimmu.2018.00101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Seliger B., Massa C. Immune therapy resistance and immune escape of tumors. Cancers (Basel) 2021;13 doi: 10.3390/cancers13030551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Xia L., Tian E., Yu M., Liu C., Shen L., Huang Y., et al. RORγt agonist enhances anti-PD-1 therapy by promoting monocyte-derived dendritic cells through CXCL10 in cancers. J Exp Clin Cancer Res. 2022;41 doi: 10.1186/s13046-022-02289-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Meijer F.A., Oerlemans G.J.M., Brunsveld L. Orthosteric and allosteric dual targeting of the nuclear receptor RORγt with a bitopic ligand. ACS Chem Biol. 2021;16:510–519. doi: 10.1021/acschembio.0c00941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Huang Y., Nahar S., Nakagawa A., Fernandez-Barrena M.G., Mertz J.A., Bryant B.M., et al. Regulation of GLI underlies a role for BET bromodomains in pancreatic cancer growth and the tumor microenvironment. Clin Cancer Res. 2016;22:4259–4270. doi: 10.1158/1078-0432.CCR-15-2068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Dickinson M., Briones J., Herrera A.F., Gonzalez-Barca E., Ghosh N., Cordoba R., et al. Phase 1b study of the BET protein inhibitor RO6870810 with venetoclax and rituximab in patients with diffuse large B-cell lymphoma. Blood Adv. 2021;5:4762–4770. doi: 10.1182/bloodadvances.2021004619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Shapiro G.I., LoRusso P., Dowlati A., K T.D., Jacobson C.A., Vaishampayan U., et al. A phase 1 study of RO6870810, a novel bromodomain and extra-terminal protein inhibitor, in patients with NUT carcinoma, other solid tumours, or diffuse large B-cell lymphoma. Br J Cancer. 2021;124:744–753. doi: 10.1038/s41416-020-01180-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Abruzzese M.P., Bilotta M.T., Fionda C., Zingoni A., Soriani A., Vulpis E., et al. Inhibition of bromodomain and extra-terminal (BET) proteins increases NKG2D ligand MICA expression and sensitivity to NK cell-mediated cytotoxicity in multiple myeloma cells: role of cMYC-IRF4-miR-125b interplay. J Hematol Oncol. 2016;9:134. doi: 10.1186/s13045-016-0362-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Adeegbe D.O., Liu Y., Lizotte P.H., Kamihara Y., Aref A.R., Almonte C., et al. Synergistic immunostimulatory effects and therapeutic benefit of combined histone deacetylase and bromodomain inhibition in non-small cell lung cancer. Cancer Discov. 2017;7:852–867. doi: 10.1158/2159-8290.CD-16-1020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Liu Y., Li Y., Liu S., Adeegbe D.O., Christensen C.L., Quinn M.M., et al. NK cells mediate synergistic antitumor effects of combined inhibition of HDAC6 and BET in a SCLC preclinical model. Cancer Res. 2018;78:3709–3717. doi: 10.1158/0008-5472.CAN-18-0161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Hammitzsch A., Tallant C., Fedorov O., O'Mahony A., Brennan P.E., Hay D.A., et al. CBP30, a selective CBP/p300 bromodomain inhibitor, suppresses human Th17 responses. Proc Natl Acad Sci U S A. 2015;112:10768–10773. doi: 10.1073/pnas.1501956112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Ghosh S., Taylor A., Chin M., Huang H.R., Conery A.R., Mertz J.A., et al. Regulatory T cell modulation by CBP/EP300 bromodomain inhibition. J Biol Chem. 2016;291:13014–13027. doi: 10.1074/jbc.M115.708560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Romero F.A., Murray J., Lai K.W., Tsui V., Albrecht B.K., An L., et al. GNE-781, a highly advanced potent and selective bromodomain inhibitor of cyclic adenosine monophosphate response element binding protein, binding protein (CBP) J Med Chem. 2017;60:9162–9183. doi: 10.1021/acs.jmedchem.7b00796. [DOI] [PubMed] [Google Scholar]
  • 260.Baldwin J., Michnoff C.H., Malmquist N.A., White J., Roth M.G., Rathod P.K., et al. High-throughput screening for potent and selective inhibitors of plasmodium falciparum dihydroorotate dehydrogenase. J Biol Chem. 2005;280:21847–21853. doi: 10.1074/jbc.M501100200. [DOI] [PubMed] [Google Scholar]
  • 261.Osiri M., Shea B., Robinson V., Suarez-Almazor M., Strand V., Tugwell P., et al. Leflunomide for treating rheumatoid arthritis. Cochrane Database Syst Rev. 2003;2002 doi: 10.1002/14651858.CD002047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Palmer A.M. Teriflunomide, an inhibitor of dihydroorotate dehydrogenase for the potential oral treatment of multiple sclerosis. Curr Opin Investig Drugs. 2010;11:1313–1323. [PubMed] [Google Scholar]
  • 263.Christian S., Merz C., Evans L., Gradl S., Seidel H., Friberg A., et al. The novel dihydroorotate dehydrogenase (DHODH) inhibitor BAY 2402234 triggers differentiation and is effective in the treatment of myeloid malignancies. Leukemia. 2019;33:2403–2415. doi: 10.1038/s41375-019-0461-5. [DOI] [PubMed] [Google Scholar]
  • 264.Sykes D.B., Kfoury Y.S., Mercier F.E., Wawer M.J., Law J.M., Haynes M.K., et al. Inhibition of dihydroorotate dehydrogenase overcomes differentiation blockade in acute myeloid leukemia. Cell. 2016;167 doi: 10.1016/j.cell.2016.08.057. 171–86.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.White R.M., Cech J., Ratanasirintrawoot S., Lin C.Y., Rahl P.B., Burke C.J., et al. DHODH modulates transcriptional elongation in the neural crest and melanoma. Nature. 2011;471:518–522. doi: 10.1038/nature09882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Echizenya S., Ishii Y., Kitazawa S., Tanaka T., Matsuda S., Watanabe E., et al. Discovery of a new pyrimidine synthesis inhibitor eradicating glioblastoma-initiating cells. Neuro Oncol. 2020;22:229–239. doi: 10.1093/neuonc/noz170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Li L., Ng S.R., Colon C.I., Drapkin B.J., Hsu P.P., Li Z., et al. Identification of DHODH as a therapeutic target in small cell lung cancer. Sci Transl Med. 2019;11 doi: 10.1126/scitranslmed.aaw7852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Olsen T.K., Dyberg C., Embaie B.T., Alchahin A., Milosevic J., Ding J., et al. DHODH is an independent prognostic marker and potent therapeutic target in neuroblastoma. JCI Insight. 2022;7 doi: 10.1172/jci.insight.153836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Ladds M., van Leeuwen I.M.M., Drummond C.J., Chu S., Healy A.R., Popova G., et al. A DHODH inhibitor increases p53 synthesis and enhances tumor cell killing by p53 degradation blockage. Nat Commun. 2018;9 doi: 10.1038/s41467-018-03441-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Mao C., Liu X., Zhang Y., Lei G., Yan Y., Lee H., et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 2021;593:586–590. doi: 10.1038/s41586-021-03539-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Colligan S.H., Amitrano A.M., Zollo R.A., Peresie J., Kramer E.D., Morreale B., et al. Inhibiting the biogenesis of myeloid-derived suppressor cells enhances immunotherapy efficacy against mammary tumor progression. J Clin Invest. 2022;132 doi: 10.1172/JCI158661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Brinkmann V., Davis M.D., Heise C.E., Albert R., Cottens S., Hof R., et al. The immune modulator FTY720 targets sphingosine 1-phosphate receptors. J Biol Chem. 2002;277:21453–21457. doi: 10.1074/jbc.C200176200. [DOI] [PubMed] [Google Scholar]
  • 273.Graler M.H., Goetzl E.J. The immunosuppressant FTY720 down-regulates sphingosine 1-phosphate G-protein-coupled receptors. FASEB J. 2004;18:551–553. doi: 10.1096/fj.03-0910fje. [DOI] [PubMed] [Google Scholar]
  • 274.Pham T.H., Okada T., Matloubian M., Lo C.G., Cyster J.G. S1P1 receptor signaling overrides retention mediated by G alpha i-coupled receptors to promote T cell egress. Immunity. 2008;28:122–133. doi: 10.1016/j.immuni.2007.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Salinas N.R., Oshima C.T., Cury P.M., Cordeiro J.A., Bueno V. FTY720 and lung tumor development. Int Immunopharmacol. 2009;9:689–693. doi: 10.1016/j.intimp.2008.12.007. [DOI] [PubMed] [Google Scholar]
  • 276.Tonelli F., Lim K.G., Loveridge C., Long J., Pitson S.M., Tigyi G., et al. FTY720 and (S)-FTY720 vinylphosphonate inhibit sphingosine kinase 1 and promote its proteasomal degradation in human pulmonary artery smooth muscle, breast cancer and androgen-independent prostate cancer cells. Cell Signal. 2010;22:1536–1542. doi: 10.1016/j.cellsig.2010.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Bayarkhangai B., Noureldin S., Yu L., Zhao N., Gu Y., Xu H., et al. A comprehensive and perspective view of oncoprotein SET in cancer. Cancer Med. 2018;7:3084–3094. doi: 10.1002/cam4.1526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Long G.V., Dummer R., Hamid O., Gajewski T.F., Caglevic C., Dalle S., et al. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. Lancet Oncol. 2019;20:1083–1097. doi: 10.1016/S1470-2045(19)30274-8. [DOI] [PubMed] [Google Scholar]

Articles from Acta Pharmaceutica Sinica. B are provided here courtesy of Elsevier

RESOURCES