Skip to main content

Interaction of telmisartan and related sartans with the programmed cell death-ligand 1 (PD-L1) protein dimer: a molecular docking analysis

Abstract

Background

Telmisartan (TLT) is a prototypic angiotensin receptor blocker largely used to treat hypertension worldwide. In addition to its cardioprotective effects, TLT presents pleiotropic activities and notably displays noticeable anti-inflammatory and antitumor effects. The repression of the programmed cell death-1 (PD-1)/programmed death-ligand 1 (PD-L1) immune checkpoint may be implicated antitumor action of TLT, as it is the case with many other compounds equipped with a biphenyl moiety. We have used molecular modeling to compare the interaction of TLT and derivatives with the PD-L1 dimer protein.

Results

Two molecules, TLT-dimer and TLT-acylglucuronide, were found to form more stable complexes with PD-L1 than TLT itself. In parallel, the docking analysis performed with a series of 12 sartans led to the identification of Olmesartan as a potential PD-L1 binder. The stacked biphenyl unit of Olmesartan positions the molecule along the groove delimited by the two protein monomers. The flanking tetrazole and imidazole moieties, on each side of the biphenyl unit of Olmesartan, contribute favorably to the protein interaction via specific hydrogen bonding interactions.

Conclusions

The computational analysis suggests a possible binding of Olmesartan to PD-L1 dimer and thus offers novel perspectives for the design of small molecules capable of interrupting the PD-1/PD-L1 immune checkpoint. Experimental studies are warranted to validate the hypothesis.

Graphical abstract

Background

Telmisartan (TLT) is one of the most frequently prescribed angiotensin receptor blockers (ARB), selectively inhibiting angiotensin II type 1 receptor (AT1) [1]. It is a classical antihypertensive drug with an excellent safety and pharmacokinetic profile, largely used to reduce arterial blood pressure in patients with hypertension, metabolic syndrome, and those at high cardiovascular risk. TLT is safe and inexpensive, used worldwide, both in Asian and Caucasian populations [2]. The global TLT market size reached US$ 3.6 billion in 2022 and continues to grow; it is expected to reach US$ 4.6 billion by 2030 [3].

This lipophilic drug is often combined with diuretics, notably hydrochlorothiazide, for the management of hypertension, affording a well-tolerated combination to treat patients with mild-to-severe hypertension [4, 5]. The water solubility (9.9 μg/mL) and bioavailability (40–60%) of TLT represent limiting factors but these properties can be improved by reducing the crystal particle size (so as to increase the surface area) and via other options, such as the development of co-amorphous formulations [6, 7].

Through its action on the renin-angiotensin system, TLT has positive effects on lipid and glucose metabolism. It is considered a drug of interest to treat non-alcoholic fatty liver disease (NAFLD) [8]. In addition to blocking angiotensin receptor, TLT has a partial peroxisome proliferator-activated receptor γ (PPARγ)-agonistic effect, which is a useful property to combat diabetes mellitus [9]. In fact, TLT has been shown to exhibit an insulin secretagogue activity, independent of AT1 receptor and PPARγ [10]. Moreover, the compound displays marked anti-inflammatory effects and is also considered a drug of interest to provide protection against Alzheimer's disease [11]. Clearly, TLT displays pleiotropic effects [12, 13].

TLT belongs to a large group of ARBs which includes a dozen of compounds (Fig. 1) with a common pharmacophore structure, but the therapeutic effectiveness of these “sartans” differs one from another [14, 15]. Irbesartan presents a higher bioavailability compared to TLT, but a shorter plasma half-life. Olmesartan displays a lower bioavailability but a higher Tmax (time to maximum plasma concentration). TLT is considered a long-acting sartan whereas Losartan equipped with a tetrazole unit exhibits a medium duration of action. EXP3174 corresponds to the active metabolite of Losartan [16]. Candesartan, Eprosartan, Azilsartan and other analogues (Fig. 1) exhibit specific safety and efficacy profiles [17].

Fig. 1
figure 1

Structures of the sartan compounds tested here

Abundant pharmacological effects have been reported with these sartan products, not limited to cardiovascular effects [15]. In particular, TLT has revealed marked antitumor effects in different models. Recently, the drug was shown to suppress tumor growth in an orthotopic transplant mouse model of glioblastoma, blocking proliferation, migration, and invasion of cancer cells [18, 19]. TLT has demonstrated anticancer activities in different models and cell lines, including prostate, renal, breast and gastric cancers [20,21,22,23,24,25] and other cancers when the drug is used alone or in combination with targeted therapy or cytotoxic drugs [26,27,28]. Different mechanisms have been invoked to account for the anticancer effects of TLT, such as a down-regulation of the transcription factor Sox9 [19], the regulation of the epithelial-to-mesenchymal transition (EMT) via down-regulation of the transcription factor genes Snail and Slug [27], antagonist targeting of N-cadherin [29] and other mechanisms. Interestingly, it has been observed that TLT can modulate activity of the immune checkpoint PD-1/PD-L1 (Programmed Death (Ligand) 1). It has been demonstrated that the expression of PD-L1 promoted in patients with obesity and metabolic syndrome could be restored by TLT [30]. More recently, it has been demonstrated that TLT combined with the kinase inhibitor osimertinib reduced PD-L1 expression in non-small cell lung cancer tissues [28]. Losartan also revealed an anticancer activity in experimental models of glioblastoma and promoted the activity of an anti-PD1 immunotherapy [31].

Immune checkpoint blockade therapies that target the programmed cell death ligand-1 (PD-L1) or its receptor programmed cell death-1 (PD-1) have revolutionized the treatment of cancers, at least for a number of solid tumors such as melanoma, lung cancer, and renal cancer. Monoclonal antibodies directed against PD-1 or PD-L1 are used to restore the antitumor response of cytotoxic T cells [32]. PD-L1 plays also a role in DNA damage repair [33, 34]. Several antibodies targeting PD-L1 (atezolizumab, avelumab, durvalumab) are already used to treat cancers but new drugs and strategies are needed to reinforce efficacy notably through the development of combination therapies and drug delivery systems [35]. In this context, small molecules targeting PD-L1 are actively searched [36]. Orally available anticancer small molecules that bind to PD-L1 and induce its dimerization have been discovered [37,38,39,40]. A few small molecule inhibitors of PD-L1, such as INCB086550, are currently undergoing clinical trials in patients with advanced solid tumors [41]. New drugs targeting PD-L1 are actively searched [42, 43].

The PD-1/PD-L1 checkpoint-associated effects of TLT may be totally indirect, not due to a drug binding to the ligand or its receptor. However, we noticed that most of these sartan compounds possess a biphenyl scaffold as found in many PD-L1-binding small molecules. Biphenyl-based small molecules are intensely studied as antitumor PD-L1 inhibitors [44,45,46]. The biphenyl unit originates from the first PD-L1 binders discovered by Bristol-Myers Squibb, such as compound BMS-202 which binds tightly to and induces dimerization of PD-L1 [47,48,49]. Over the past seven years, numerous biphenyl-containing molecules and hybrid compounds targeting PD-L1 have been designed [50,51,52,53]. A biphenyl core is also included in PD-L1 positron emission tomography tracers [54, 55]. It can be found in other drugs which can be combined with monoclonal antibodies targeting PD-1 or PD-L1. For example, the biphenyl-containing drug tazemetostat was shown to combine well with anti-PD-L1 atezolizumab in lymphoma patients [56].

These considerations prompted us to investigate the potential interaction of sartan compounds with the PD-L1 protein using a molecular docking approach, starting from the crystal structure of PD-L1 bound BMS-202 [47]. We have previously used the same approach to identify other PD-L1-binding molecules containing a biphenyl scaffold [57]. Here we examined the potential interaction of TLT, its metabolites and analogous sartan molecules with PD-L1.

Methods

In silico molecular docking procedure

The tridimensional structure of the dimeric form of the extracellular domain of PD-L1 was retrieved from the Protein Data Bank (www.rcsb.org) under the PDB code 5J89 [58]. The GOLD 5.3 software (Cambridge Crystallographic Data Centre, Cambridge, UK) was used to perform molecular docking analysis. Prior to the docking operations, the structure of each ligand was optimized using a classical Monte Carlo conformational searching procedure via the BOSS software [59]. Molecular graphics and analysis were performed using Discovery Studio Visualizer, Biovia 2020 (Dassault Systèmes BIOVIA Discovery Studio Visualizer 2020, San Diego, Dassault Systèmes, 2020).

The PD-L1 protein structure (5J89) includes the biphenyl small molecule BMS-202 bound to the interface of two face-to-face monomers. The BMS-202 binding site was considered as the potential binding site for the studied sartan compounds. During the process, the side chains of the following amino acids within the binding site were rendered fully flexible: Tyr56, Met115, Asp122, Tyr123, and Lys124 (monomer A), and Tyr56, Gln66, Met115, Asp122, and Tyr123 (monomer B). A docking grid centered in the volume defined by the central amino acid has been defined based on shape complementarity and geometry considerations. In general, up to 100 poses considered as energetically reasonable are selected during the search for the correct binding mode of the ligand. The decision to select a trial pose is based on ranked poses, using the fitness scoring function (PLP score) [60]. The same procedure was used to establish molecular models for all sartan compounds.

The Boss program and the Molecular Mechanics/Generalized Born Surface Area (MM/GBSA) procedure were used to evaluate free energies of hydration (ΔG, also designated Δμh [61] or HFE [62]), in relation with aqueous solubility [63]. The Boss program was also used to evaluate the stability of the receptor-ligand complex through the empirical potential energy of interaction (ΔE) [64, 65]. The empirical potential energy of interaction ΔE calculated for each drug-protein complex was defined using the equation ΔE(interaction) = E(complex)—(E(protein) + E(ligand)), using the Spectroscopic Empirical Potential Energy function SPASIBA [64, 65]. SPASIBA has been specifically developed to provide refined empirical molecular mechanics force field parameters, as described in other studies [64, 66]. Using this specific force field for, Monte Carlo (MC) simulations achieve the same level of convergence as Molecular Dynamics (MD), with less computer time [67].

Results

Docking interactions of TLT and its metabolites with PD-L1

We started our analysis using the structure of the reference compound BMS-202 bound to recombinant PD-L1 (PDB: 5J89) [58]. The drug binds to the interface of two monomers, stabilizing a dimeric form of the protein and defining an extended cavity that can be exploited for drug binding [47]. TLT was docked into the cavity and its capacity to form stable complexes with PD-L1 was evaluated though the calculation of the empirical energy of interaction (ΔE), as reported in Table 1. The analysis indicated that TLT can form complexes with PD-L1 but the calculated ΔE value was superior (less negative) compared to that measured with BMS-202, suggesting a weaker affinity. The two compounds are comparable in term of hydration free energy (ΔG). TLT is an acid which is a little more favorably hydrated than the reference BMS-202 (Table 1). In general, the free energies of hydration for acidic residues are more favorable than for basic residues [61].

Table 1 Calculated potential energy of interaction (ΔE) and free energy of hydration (ΔG) for the interaction of TLT and derivatives with PD-L1

Different derivatives of TLT were then tested, such as the amide and methyl ester forms but no improvement was observed for these two compounds. The two derivatives TLT-terbutyl ester and TLT- N-desmethyl gave slightly better results with a ΔE around − 80 kcal/mol, comparable to the value measured with BMS-202. A further improvement of the PD-L1 interaction was observed with two other molecules: a dimeric compound and a glucuronide derivative of TLT (Fig. 1). The TLT dimeric compound corresponds to a minor impurity detected in TLT tablets [68]. This compound has no biological relevance, but the observation suggests that an extension of the drug structure could reinforce drug binding to PD-L1. With this dimer, the drug-protein interaction is stabilized via a variety of van der Walls contacts and π-stacking interactions. But a large portion of the elongated molecule protrudes outside the binding cavity, as shown in Fig. 2. The molecule is too long, not perfectly adapted to the binding surface but nevertheless it offers a linear extended part that inserts well into the protein interface.

Fig. 2
figure 2

Molecular model of the TLT dimer bound to the dimeric form of PD-L1. a The TLT-dimer compound bound at the interface of the two PD-L1 units (in cyan and green). b A close-up view of the PD-L1-bound ligand with the solvent-accessible surface (SAS) surrounding the drug binding zone (color code indicated). c Binding map contacts for TLT-dimer bound to PD-L1 (color code indicated)

The case of TLT-acylglucuronide derivative (Fig. 1) is more interesting because it corresponds to a major phase II liver metabolite of TLT [69]. It is an inactive elimination product, formed in the liver and excreted through the hepatobiliary system but an intestinal deconjugation of the TLT glucuronide metabolite restoring the parent compound via the enterohepatic recirculation can occur [70]. We found that the compound TLT-glucuronide has a capacity to form very stable complexes with PD-L1, with an empirical potential energy of interaction ΔE of − 100 kcal/mol calculated for this glycosyl conjugate bound to the interface of the PD-L1 dimer. In this case, the acyl-β-D-glucuronide moiety projects toward the concave (inward) face of the dimeric protein structure and contributes significantly to the protein interaction via two hydrogen bonds with residues Tyr123 and Lys124, whereas the two benzimidazole units are inserted into the narrow protein groove in the hydrophobic part of the channel (Fig. 3). This 1-O-acylglucuronide of TLT is a stable circulating product, with a low binding to human serum albumin, but it is rapidly cleared (clearance of 180 ml/min/kg compared with 15.6 ml/min/kg for TLT), resulting in a low systemic exposure [71]. Therefore, this TLT-glucuronide may not contribute to blocking PD-L1 but here again, the information is important in terms of drug design. The substitution of the acid function of TLT on the biphenyl portion apparently represents a suitable option to obtain novel PD-L1 binders.

Fig. 3
figure 3

Molecular model of TLT-glucuronide bound to the dimeric form of PD-L1. a The TLT-acylglucuronide compound bound at the interface of the two PD-L1 units (in cyan and green). b A close-up view of the PD-L1-bound ligand with the hydrophobicity surrounding the drug binding zone (color code indicated). c Binding map contacts for TLT-glucuronide bound to PD-L1 (color code as in Fig. 2)

Docking interactions of other sartans with PD-L1

The modeling analysis was extended to a series of 12 sartans, including close analogues of TLT such as the benzimidazole Pomisartan and different analogues bearing a tetrazole unit attached to the biphenyl core, such as Candesartan and Valsartan. For each compound, molecular models were constructed and their potential interaction with PD-L1 was evaluated through the calculations of the empirical energy of interaction (ΔE) and energy of hydration (ΔG or hydration free energy HFE). The values are collated in Table 2. Binding maps are shown in Additional file 1: Fig. S1. No profound improvement was observed compared to TLT. The weaker binder was the imidazolinone derivative Irbesartan and the best compound was the imidazole derivative Olmesartan, but all compounds gave ΔE values higher (less negative) than that calculated with the reference BMS-202. Losartan active metabolite EXP3174 emerged as a poor binder, as it is the case with Enoltasosartan.

Table 2 Calculated potential energy of interaction (ΔE) and free energy of hydration (ΔG) for the interaction of selected sartans with PD-L1

The investigation was extended to search for other compounds susceptible to bind to PD-L1, but no compound better than Olmesartan was identified. For examples, we tested derivatives of the AT1 antagonist Eprosartan (lacking a biphenyl unit) which has been shown recently to exhibit antioxidative and anti-inflammatory properties [72] but we found no improved binding when testing Eprosartan (CID: 5281037; ΔE = − 66.85 kcal/mol), methyl-Eprosartan (CID: 45358786; ΔE = − 75.40 kcal/mol), and ethyl-Eprosartan (CID: 10049536; ΔE = − 79.80 kcal/mol).

Among these sartan compounds, the best molecule for interacting with PD-L1 is Olmesartan with its biphenyl unit stacking over residue Tyr56 and its tetrazole unit H-bonding to Asp122. It is interesting to note that the other side of the molecule is also well engaged in the protein interaction, with the 2-OH group on the propyl-imidazole moiety implicated in two vicinal H-bonds with Ala18 and Phe19. Olmesartan emerges as a potential PD-L1 binder. The best binding pose selected with Olmesartan is not ideal because there is only a van der Walls contact (a weak attraction) between the biphenyl unit and residue Tyr123, in addition to the essential stacking interaction with residue Tyr56. There exists an alternative pose, less favorable in terms of computed energies (ΔE = − 70.00 kcal/mol and ΔG = − 18.60 kcal/mol), but characterized by a stacking interaction between the tetrazole unit of Olmesartan and Tyr123 (Additional file 1: Fig. S2).

Discussion

The established anticancer activity of TLT and the presence of a biphenyl unit in the drug structure prompted us to investigate the potential binding of TLT and derivatives to the immune checkpoint ligand PD-L1. The modeling analysis suggests that drugs like TLT and Olmesartan could interact with PD-L1, thus possibly playing a role in their antitumor action. The hypothesis remains weak at present but there are interesting elements to consider. A moderate reduced cancer-specific mortality has been noted among users of angiotensin receptor blockers (ARB) [73]. ARB seem to exhibit a significant overall protective against lung, bladder and colorectal cancers [74]. The anticancer activity of TLT has been well characterized in different experimental tumor models and its analogue Olmesartan has been shown to exert an antitumor action, notably in pancreatic cancer, and cervical cancer through an upregulation of microRNA miR-205 and inhibition of VEGF-A expression [75,76,77,78]. It displays also a significant anti-inflammatory action [79]. This biphenyl drug was shown to potentiate the anti-angiogenic effect of sorafenib [80]. Irbesartan has been shown also to repress the proliferation of cancer cells [81] and to overcome chemoresistance [82]. Therefore, there are good reasons to investigate further the antitumor potential and immune effects of these sartan drugs. Very interestingly, Irbesartan has been found to activate an immune response and in particular the infiltration of CD8+ T cells in relapsed tumors [83]. ARB can facilitate tumor infiltration by effector T cells [84]. It is therefore conceivable that ARB can modulate the PD-1/PD-L1 checkpoint (Fig. 4).

Fig. 4
figure 4

Molecular model of the Olmesartan bound to the dimeric form of PD-L1. a Olmesartan bound at the interface of the two PD-L1 units (in cyan and green). b A close-up view of the PD-L1-bound ligand with the solvent-accessible surface (SAS) surrounding the drug binding zone (color code indicated). c Binding map contacts for Olmesartan bound to PD-L1 dimer (color code as in Fig. 2)

Here we observed that Olmesartan is better adapted than TLT for binding to PD-L1. Olmesartan is perhaps not the best sartan to study because it can induce digestive tract injuries (parenthetically a side effects commonly observed also with immune checkpoint inhibitors). However, the observations further attest of the benefit of considering diverse biphenyl compounds has potential PD-L1 binders. Recent studies have underlined the possibility to affect the functionality of the PD-1/PD-L1 checkpoint with diverse biphenyl or biaryl compounds and the great benefits of computational approaches to identify novel PD-L1 binders [85,86,87,88,89]. Our study brings another brick in the wall, suggesting to consider further some of the sartan compounds as potential modular of the PD-L1 checkpoint. The mode of binding of these sartans at the interface of the PD-L1 dimer is similar to that observed with the reference ligand BMS-202 (Fig. 5).

Fig. 5
figure 5

Molecular model of the PD-L1 dimer stabilized with TLT (magenta) or the reference ligand BMS-202 (yellow), with the two molecules superimposed at the junction between the two PD-L1 monomers. The chemical structures of the two molecules are shown

In conclusion, our molecular docking analysis has identified the drug Olmesartan as a potential binder to the immune checkpoint protein PD-L1. The drug has the capacity to interact with the PD-L1 dimer, via its biphenyl core. The study provides guidance for the design of novel PD-L1 binders, based on the structure of diverse sartan compounds.

Availability of data and materials

Not applicable (no data).

Abbreviations

ARB:

Angiotensin receptor blockers

AT1:

Angiotensin II type 1 receptor

PPARγ:

Peroxisome proliferator-activated receptor γ

PD-1:

Programmed cell death-1

PD-L1:

Programmed death-ligand 1

TLT:

Telmisartan

References

  1. Abdelkader NN, Awaisu A, Elewa H, El Hajj MS (2023) Prescribing patterns of antihypertensive medications: a systematic review of literature between 2010 and 2020. Explor Res Clin Soc Pharm 11:100315

    PubMed  PubMed Central  Google Scholar 

  2. Kumari K, Toppo MS, Majhi L, Kumar A (2022) Blood pressure-lowering effect of telmisartan compared to losartan among mild to moderate essential hypertensive adult subjects: a meta-analysis. J Family Med Prim Care 11:6227–6235

    Article  PubMed  PubMed Central  Google Scholar 

  3. Telmisartan market (2022) https://straitsresearch.com/report/telmisartan-market

  4. Kjeldsen S, Mancia G, Schmieder R, Mattheus M, Unger T (2013) An update on telmisartan/hydrochlorothiazide combinations for the management of hypertensive patients with additional cardiovascular risk factors. Expert Rev Cardiovasc Ther 11:673–682

    Article  CAS  PubMed  Google Scholar 

  5. Kjeldsen SE, Schumacher H, Neldam S, Guthrie RM (2013) Telmisartan/Hydrochlorothiazide combination therapy for the treatment of hypertension: a pooled analysis in older and younger patients. J Clin Hypertens (Greenwich) 15:380–388

    Article  CAS  PubMed  Google Scholar 

  6. Almotairy A, Almutairi M, Althobaiti A, Alyahya M, Sarabu S, Alzahrani A, Zhang F, Bandari S, Repka MA (2021) Effect of pH modifiers on the solubility, dissolution rate, and stability of telmisartan solid dispersions produced by hot-melt extrusion technology. J Drug Deliv Sci Technol 65:102674

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hatanaka Y, Uchiyama H, Kaneko S, Ueda K, Higashi K, Moribe K, Furukawa S, Takase M, Yamanaka S, Kadota K, Tozuka Y (2023) Designing a novel coamorphous salt formulation of telmisartan with amlodipine to enhance permeability and oral absorption. Mol Pharm 20:4071–4085

    Article  CAS  PubMed  Google Scholar 

  8. Borém LMA, Neto JFR, Brandi IV, Lelis DF, Santos SHS (2018) The role of the angiotensin II type I receptor blocker telmisartan in the treatment of non-alcoholic fatty liver disease: a brief review. Hypertens Res 41:394–405

    Article  PubMed  PubMed Central  Google Scholar 

  9. Ayza MA, Zewdie KA, Tesfaye BA, Gebrekirstos ST, Berhe DF (2020) Anti-diabetic effect of telmisartan through its partial PPARγ-agonistic activity. Diabetes Metab Syndr Obes 13:3627–3635

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Liu T, Cui L, Xue H, Yang X, Liu M, Zhi L, Yang H, Liu Z, Zhang M, Guo Q, He P, Liu Y, Zhang Y (2021) Telmisartan potentiates insulin secretion via ion channels, independent of the AT1 receptor and PPARγ. Front Pharmacol 12:739637

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Fu XX, Wei B, Cao HM, Duan R, Deng Y, Lian HW, Zhang YD, Jiang T (2023) Telmisartan alleviates Alzheimer’s disease-related neuropathologies and cognitive impairments. J Alzheimers Dis 94:919–933

    Article  CAS  PubMed  Google Scholar 

  12. Rizos CV, Elisaf MS, Liberopoulos EN (2009) Are the pleiotropic effects of telmisartan clinically relevant? Curr Pharm Des 15:2815–2832

    Article  CAS  PubMed  Google Scholar 

  13. Vitiello A, La Porta R, Trama U, Troiano V, Ferrara F (2022) Pleiotropic effects of AT-1 receptor antagonists in hypoxia induced by cardiac ischaemia. Inflammopharmacol 30:1407–1410

    Article  CAS  Google Scholar 

  14. Muszalska I, Sobczak A, Dołhań A, Jelińska A (2014) Analysis of Sartans: a review. J Pharm Sci 103:2–28

    Article  CAS  PubMed  Google Scholar 

  15. Wang YF, Ren XY, Zhang W, Rao GW (2023) Research progress in pharmacological mechanisms, structure-activity relationship and synthesis of sartans. Curr Med Chem 30:2247–2266

    Article  CAS  PubMed  Google Scholar 

  16. Wani TU, Mir KB, Raina A, Dar AA, Jan I, Khan NA, Wani TA, Sofi JA, Hassan GI, Almoallim HS, Alharbi SA, Ansari MJ, Alfarraj S, Tarique M, Dar SA (2023) Simultaneous quantification of losartan potassium and its active metabolite, EXP3174, in rabbit plasma by validated HPLC-PDA. Biomed Chromatogr 37:e5645

    Article  CAS  PubMed  Google Scholar 

  17. Abraham HM, White CM, White WB (2015) The comparative efficacy and safety of the angiotensin receptor blockers in the management of hypertension and other cardiovascular diseases. Drug Saf 38:33–54

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wang Y, Zhang T, Li C, Guo J, Xu B, Xue L (2022) Telmisartan attenuates human glioblastoma cells proliferation and oncogenicity by inducing the lipid oxidation. Asia Pac J Clin Oncol 18:217–223

    Article  CAS  PubMed  Google Scholar 

  19. Chang YL, Chou CH, Li YF, Huang LC, Kao Y, Hueng DY, Tsai CK (2023) Antiproliferative and apoptotic effects of telmisartan in human glioma cells. Cancer Cell Int 23:111

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Funao K, Matsuyama M, Kawahito Y, Sano H, Chargui J, Touraine JL, Nakatani T, Yoshimura R (2008) Telmisartan is a potent target for prevention and treatment in human prostate cancer. Oncol Rep 20:295–300

    CAS  PubMed  Google Scholar 

  21. Funao K, Matsuyama M, Kawahito Y, Sano H, Chargui J, Touraine JL, Nakatani T, Yoshimura R (2009) Telmisartan as a peroxisome proliferator-activated receptor-γ ligand is a new target in the treatment of human renal cell carcinoma. Mol Med Rep 2:193–198

    CAS  PubMed  Google Scholar 

  22. Fujita N, Fujita K, Iwama H, Kobara H, Fujihara S, Chiyo T, Namima D, Yamana H, Kono T, Takuma K, Hirata M, Kobayashi K, Kato K, Kamada H, Morishita A, Tsutsui K, Himoto T, Okano K, Suzuki Y, Masaki T (2020) Antihypertensive drug telmisartan suppresses the proliferation of gastric cancer cells in vitro and in vivo. Oncol Rep 44:339–348

    Article  CAS  PubMed  Google Scholar 

  23. Tsujiya Y, Hasegawa A, Yamamori M, Okamura N (2021) Telmisartan-induced cytotoxicity via G2/M phase arrest in renal cell carcinoma cell lines. Biol Pharm Bull 44:1878–1885

    Article  CAS  PubMed  Google Scholar 

  24. Tsujiya Y, Yamamori M, Hasegawa AI, Yamamoto Y, Yashiro M, Okamura N (2021) Telmisartan exerts cytotoxicity in scirrhous gastric cancer cells by inducing G0/G1 cell cycle arrest. Anticancer Res 41:5461–5468

    Article  CAS  PubMed  Google Scholar 

  25. Kumar U, Aich J, Devarajan S (2023) Exploring the repurposing potential of telmisartan drug in breast cancer: an in-silico and in-vitro approach. Anticancer Drugs 34:1094–1103

    Article  CAS  PubMed  Google Scholar 

  26. Arthur P, Patel N, Surapaneni SK, Mondal A, Gebeyehu A, Bagde A, Kutlehria S, Nottingham E, Singh M (2020) Targeting lung cancer stem cells using combination of Tel and Docetaxel liposomes in 3D cultures and tumor xenografts. Toxicol Appl Pharmacol 401:115112

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Khorsand M, Mostafavi-Pour Z, Razban V, Khajeh S, Zare R (2022) Combinatorial effects of telmisartan and docetaxel on cell viability and metastatic gene expression in human prostate and breast cancer cells. Mol Biol Res Commun 11:11–20

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Nimma R, Kalvala AK, Patel N, Surapaneni SK, Sun L, Singh R, Nottingham E, Bagde A, Kommineni N, Arthur P, Nathani A, Meckes DG Jr, Singh M (2022) Combined transcriptomic and proteomic profiling to unravel osimertinib, CARP-1 functional mimetic (CFM 4.17) formulation and telmisartan combo treatment in NSCLC tumor xenografts. Pharmaceutics 14:1156

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Khorsand M, Khajeh S, Eslami M, Nezafat N, Ghasemi Y, Razban V, Mostafavi-Pour Z (2022) Telmisartan anti-cancer activities mechanism through targeting N-cadherin by mimicking ADH-1 function. J Cell Mol Med 26:2392–2403

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wang Y, Zhang X, Xie X, Chen W, Li M, Diao D, Dang C (2020) Obesity and metabolic syndrome related macrophage promotes PD-L1 expression in TNBC through IL6/JAK/STAT pathway and can be reversed by telmisartan. Cancer Biol Ther 21:1179–1190

    Article  PubMed  PubMed Central  Google Scholar 

  31. Datta M, Chatterjee S, Perez EM, Gritsch S, Roberge S, Duquette M, Chen IX, Naxerova K, Kumar AS, Ghosh M, Emblem KE, Ng MR, Ho WW, Kumar P, Krishnan S, Dong X, Speranza MC, Neagu MR, Iorgulescu JB, Huang RY, Youssef G, Reardon DA, Sharpe AH, Freeman GJ, Suvà ML, Xu L, Jain RK (2023) Losartan controls immune checkpoint blocker-induced edema and improves survival in glioblastoma mouse models. Proc Natl Acad Sci USA 120:e2219199120

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Philips GK, Atkins M (2015) Therapeutic uses of anti-PD-1 and anti-PD-L1 antibodies. Int Immunol 27:39–46

    Article  CAS  PubMed  Google Scholar 

  33. Zhang T, Zheng S, Liu Y, Li X, Wu J, Sun Y, Liu G (2021) DNA damage response and PD-1/PD-L1 pathway in ovarian cancer. DNA Repair (Amst) 102:103112

    Article  CAS  PubMed  Google Scholar 

  34. Zhou Z, Wang H, Li J, Jiang X, Li Z, Shen J (2023) Recent progress, perspectives, and issues of engineered PD-L1 regulation nano-system to better cure tumor: A review. Int J Biol Macromol 254:127911

    Article  PubMed  Google Scholar 

  35. Yin S, Chen Z, Chen D, Yan D (2023) Strategies targeting PD-L1 expression and associated opportunities for cancer combination therapy. Theranostics 13:1520–1544

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zwergel C, Fioravanti R, Mai A (2023) PD-L1 small-molecule modulators: A new hope in epigenetic-based multidrug cancer therapy? Drug Discov Today 28:103435

    Article  CAS  PubMed  Google Scholar 

  37. Guo Y, Jin Y, Wang B, Liu B (2021) Molecular mechanism of small-molecule inhibitors in blocking the PD-1/PD-L1 pathway through PD-L1 dimerization. Int J Mol Sci 22:4766

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Liu C, Zhou F, Yan Z, Shen L, Zhang X, He F, Wang H, Lu X, Yu K, Zhao Y, Zhu D (2021) Discovery of a novel, potent and selective small-molecule inhibitor of PD-1/PD-L1 interaction with robust in vivo anti-tumour efficacy. Br J Pharmacol 178:2651–2670

    Article  CAS  PubMed  Google Scholar 

  39. Wang F, Ye W, Wang S, He Y, Zhong H, Wang Y, Zhu Y, Han J, Bing Z, Ji S, Liu H, Yao X (2021) Discovery of a new inhibitor targeting PD-L1 for cancer immunotherapy. Neoplasia 23:281–293

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hao X, Chen Z, Li H, Wei M, Zuo Z, Su Q (2023) Small-Molecule Drugs in Immunotherapy. Mini Rev Med Chem 23:1341–1359

    Article  CAS  PubMed  Google Scholar 

  41. Koblish HK, Wu L, Wang LS, Liu PCC, Wynn R, Rios-Doria J, Spitz S, Liu H, Volgina A, Zolotarjova N, Kapilashrami K, Behshad E, Covington M, Yang YO, Li J, Diamond S, Soloviev M, O’Hayer K, Rubin S, Kanellopoulou C, Yang G, Rupar M, DiMatteo D, Lin L, Stevens C, Zhang Y, Thekkat P, Geschwindt R, Marando C, Yeleswaram S, Jackson J, Scherle P, Huber R, Yao W, Hollis G (2022) Characterization of INCB086550: a potent and novel small-molecule PD-L1 inhibitor. Cancer Discov 12:1482–1499

    Article  PubMed  PubMed Central  Google Scholar 

  42. Thuru X, Magnez R, El-Bouazzati H, Vergoten G, Quesnel B, Bailly C (2022) Drug repurposing to enhance antitumor response to PD-1/PD-L1 immune checkpoint inhibitors. Cancers (Basel) 14:3368

    Article  CAS  PubMed  Google Scholar 

  43. Chandrasekaran J, Elumalai S, Murugesan V, Kunjiappan S, Pavadai P, Theivendren P (2023) Computational design of PD-L1 small molecule inhibitors for cancer therapy. Mol Divers 27:1633–1644

    Article  CAS  PubMed  Google Scholar 

  44. Wang T, Cai S, Wang M, Zhang W, Zhang K, Chen D, Li Z, Jiang S (2021) Novel biphenyl pyridines as potent small-molecule inhibitors targeting the programmed cell death-1/programmed cell death-ligand 1 interaction. J Med Chem 64:7390–7403

    Article  CAS  PubMed  Google Scholar 

  45. Chen R, Yuan D, Ma J (2022) Advances of biphenyl small-molecule inhibitors targeting PD-1/PD-L1 interaction in cancer immunotherapy. Future Med Chem 14:97–113

    Article  CAS  PubMed  Google Scholar 

  46. Sasmal P, Kumar Babasahib S, Prashantha Kumar BR, Manjunathaiah Raghavendra N (2022) Biphenyl-based small molecule inhibitors: Novel cancer immunotherapeutic agents targeting PD-1/PD-L1 interaction. Bioorg Med Chem 73:117001

    Article  CAS  PubMed  Google Scholar 

  47. Zak KM, Grudnik P, Magiera K, Dömling A, Dubin G, Holak TA (2017) Structural biology of the immune checkpoint receptor PD-1 and its ligands PD-L1/PD-L2. Structure 25:1163–1174

    Article  CAS  PubMed  Google Scholar 

  48. Guzik K, Zak KM, Grudnik P, Magiera K, Musielak B, Törner R, Skalniak L, Dömling A, Dubin G, Holak TA (2017) Small-molecule inhibitors of the programmed cell death-1/programmed death-ligand 1 (PD-1/PD-L1) interaction via transiently induced protein states and dimerization of PD-L1. J Med Chem 60:5857–5867

    Article  CAS  PubMed  Google Scholar 

  49. Skalniak L, Zak KM, Guzik K, Magiera K, Musielak B, Pachota M, Szelazek B, Kocik J, Grudnik P, Tomala M, Krzanik S, Pyrc K, Dömling A, Dubin G, Holak TA (2017) Small-molecule inhibitors of PD-1/PD-L1 immune checkpoint alleviate the PD-L1-induced exhaustion of T-cells. Oncotarget 8:72167–72181

    Article  PubMed  PubMed Central  Google Scholar 

  50. Narva S, Xiong X, Ma X, Tanaka Y, Wu Y, Zhang W (2020) Synthesis and evaluation of biphenyl-1,2,3-triazol-benzonitrile derivatives as PD-1/PD-L1 inhibitors. ACS Omega 5:21181–21190

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. OuYang Y, Gao J, Zhao L, Lu J, Zhong H, Tang H, Jin S, Yue L, Li Y, Guo W, Xu Q, Lai Y (2021) Design, synthesis, and evaluation of o-(biphenyl-3-ylmethoxy)nitrophenyl derivatives as PD-1/PD-L1 inhibitors with potent anticancer efficacy in vivo. J Med Chem 64:7646–7666

    Article  CAS  PubMed  Google Scholar 

  52. Liu J, Cheng Y, Yuan L, Liu T, Ruan Y, Ren Y, Li L, Jiang S, Xiao Y, Chen J (2023) Discovery and crystallography study of novel biphenyl ether and oxadiazole thioether (non-arylmethylamine)-based small-molecule PD-1/PD-L1 inhibitors as immunotherapeutic agents. J Med Chem 66:13172–13188

    Article  CAS  PubMed  Google Scholar 

  53. Jing T, Zhang Z, Kang Z, Mo J, Yue X, Lin Z, Fu X, Liu C, Ma H, Zhang X, Hu W (2023) Discovery and optimization of novel biphenyl derivatives bearing cyclopropyl linkage as potent programmed cell death-1/programmed cell death-ligand 1 inhibitors. J Med Chem 66:6811–6835

    Article  CAS  PubMed  Google Scholar 

  54. Bamminger K, Pichler V, Vraka C, Nehring T, Pallitsch K, Lieder B, Hacker M, Wadsak W (2023) On the road towards small-molecule programmed cell death 1 ligand 1 positron emission tomography tracers: a ligand-based drug design approach. Pharmaceuticals (Basel) 16:1051

    Article  CAS  PubMed  Google Scholar 

  55. Xu L, Zhang L, Liang B, Zhu S, Lv G, Qiu L, Lin J (2023) Design, synthesis, and biological evaluation of a small-molecule PET agent for imaging PD-L1 expression. Pharmaceuticals (Basel) 16:213

    Article  CAS  PubMed  Google Scholar 

  56. Palomba ML, Cartron G, Popplewell L, Ribrag V, Westin J, Huw LY, Agarwal S, Shivhare M, Hong WJ, Raval A, Chang AC, Penuel E, Morschhauser F (2022) Combination of atezolizumab and tazemetostat in patients with relapsed/refractory diffuse large B-cell lymphoma: results from a phase Ib study. Clin Lymphoma Myeloma Leuk 22:504–512

    Article  CAS  PubMed  Google Scholar 

  57. Bailly C, Vergoten G (2020) Flurbiprofen as a biphenyl scaffold for the design of small molecules binding to PD-L1 protein dimer. Biochem Pharmacol 178:114042

    Article  CAS  PubMed  Google Scholar 

  58. Zak KM, Grudnik P, Guzik K, Zieba BJ, Musielak B, Dömling A, Dubin G, Holak TA (2016) Structural basis for small molecule targeting of the programmed death ligand 1 (PD-L1). Oncotarget 7:30323–30335

    Article  PubMed  PubMed Central  Google Scholar 

  59. Jorgensen WL, Tirado-Rives J (2005) Molecular modeling of organic and biomolecular systems using BOSS and MCPRO. J Comput Chem 26:1689–1700

    Article  CAS  PubMed  Google Scholar 

  60. Jones G, Willett P, Glen RC, Leach AR, Taylor R (1997) Development and validation of a genetic algorithm for flexible docking. J Mol Biol 267:727–748

    Article  CAS  PubMed  Google Scholar 

  61. Fossat MJ, Zeng X, Pappu RV (2021) Uncovering differences in hydration free energies and structures for model compound mimics of charged side chains of amino acids. J Phys Chem B 125:4148–4161

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kinoshita M, Hayashi T (2020) Accurate and rapid calculation of hydration free energy and its physical implication for biomolecular functions. Biophys Rev 12:469–480

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Jorgensen WL, Ulmschneider JP, Tirado-Rives J (2004) Free energies of hydration from a generalized Born model and an ALL-atom force field. J Phys Chem B 108:16264–16270

    Article  CAS  Google Scholar 

  64. Vergoten G, Mazur I, Lagant P, Michalski JC, Zanetta JP (2003) The SPASIBA force field as an essential tool for studying the structure and dynamics of saccharides. Biochimie 85:65–73

    Article  CAS  PubMed  Google Scholar 

  65. Lagant P, Nolde D, Stote R, Vergoten G, Karplus M (2004) Increasing normal modes analysis accuracy: the SPASIBA spectroscopic force field introduced into the CHARMM program. J Phys Chem A 108:4019–4029

    Article  CAS  Google Scholar 

  66. Meziane-Tani M, Lagant P, Semmoud A, Vergoten G (2006) The SPASIBA force field for chondroitin sulfate: vibrational analysis of D-glucuronic and N-acetyl-D-galactosamine 4-sulfate sodium salts. J Phys Chem A 110:11359–11370

    Article  CAS  PubMed  Google Scholar 

  67. Jorgensen WL, Tirado-Rives J (1996) Monte Carlo versus molecular dynamics for conformational sampling. J Phys Chem 100:14508–14513

    Article  CAS  Google Scholar 

  68. Marineni B, Reddy TS (2014) Simultaneous determination of telmisartan impurities and chlorthalidone impurities by UPLC. Int J Pharm Sci Rev Res 26:226–230

    CAS  Google Scholar 

  69. Šilhavý J, Mlejnek P, Šimáková M, Vaněčková I, Behuliak M, Kuda O, Sticová E, Jirsa M, Pravenec M (2018) Acute toxic effects of telmisartan in spontaneously hypertensive rats fed a high fructose diet. Physiol Res 67:851–856

    Article  PubMed  Google Scholar 

  70. Li R, Ghosh A, Maurer TS, Kimoto E, Barton HA (2014) Physiologically based pharmacokinetic prediction of telmisartan in human. Drug Metab Dispos 42:1646–1655

    Article  CAS  PubMed  Google Scholar 

  71. Ebner T, Heinzel G, Prox A, Beschke K, Wachsmuth H (1999) Disposition and chemical stability of telmisartan 1-O-acylglucuronide. Drug Metab Dispos 27:1143–1149

    CAS  PubMed  Google Scholar 

  72. Lotfi B, Bagheri Y, Abdollahpour A, Ahmadian E, Matin S, Firouzfar A, Zununi Vahed S, Khajepour F (2023) Protective effect of eprosartan against ischemic acute renal injury: acting on NF-κB, caspase 3, and Sirtuin 1. Int Immunopharmacol 115:109690

    Article  CAS  PubMed  Google Scholar 

  73. Busby J, McMenamin Ú, Spence A, Johnston BT, Hughes C, Cardwell CR (2018) Angiotensin receptor blocker use and gastro-oesophageal cancer survival: a population-based cohort study. Aliment Pharmacol Ther 47:279–288

    Article  CAS  PubMed  Google Scholar 

  74. Dagher YG, El Helou S, Haifa KG, Chalhoub IG, Boulos RT, Atallah B, Nasr F, Kassab I, Chahine MN (2023) The association between angiotensin receptor blockers and lung, bladder, and colon cancer development: a 10-year multicentric retrospective Lebanese study. Medicine (Baltimore) 102:e34901

    Article  CAS  PubMed  Google Scholar 

  75. Masamune A, Hamada S, Kikuta K, Takikawa T, Miura S, Nakano E, Shimosegawa T (2013) The angiotensin II type I receptor blocker olmesartan inhibits the growth of pancreatic cancer by targeting stellate cell activities in mice. Scand J Gastroenterol 48:602–609

    Article  CAS  PubMed  Google Scholar 

  76. Bakhtiari E, Hosseini A, Boroushaki MT, Mousavi SH (2015) Synergistic, cytotoxic and apoptotic activities of olmesartan with NF-κB inhibitor against HeLa human cell line. Toxicol Mech Methods 25:614–621

    CAS  PubMed  Google Scholar 

  77. Bakhtiari E, Hosseini A, Boroushaki MT, Mousavi SH (2016) Angiotensin II receptor antagonist olmesartan and NF-kappaB inhibitor as cytotoxic and apoptotic agents in MCF-7 human cell line. J Chemother 28:314–320

    Article  CAS  PubMed  Google Scholar 

  78. Yue Z, Yun-Shan Z, Feng-Xia X (2016) miR-205 mediates the inhibition of cervical cancer cell proliferation using olmesartan. J Renin Angiotensin Aldosterone Syst 17:1470320316663327

    Article  PubMed  PubMed Central  Google Scholar 

  79. Liu Y, Liu J, Ma Y, Zhang Y, Chen Q, Yang X, Shang Y (2022) The protective effects of Olmesartan against interleukin-29 (IL-29)-induced type 2 collagen degradation in human chondrocytes. Bioengineered 13:1802–1813

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Abd-Alhaseeb MM, Zaitone SA, Abou-El-Ela SH, Moustafa YM (2014) Olmesartan potentiates the anti-angiogenic effect of sorafenib in mice bearing Ehrlich’s ascites carcinoma: role of angiotensin (1–7). PLoS ONE 9:e85891

    Article  PubMed  PubMed Central  Google Scholar 

  81. Lu YU, Miyamoto T, Takeuchi H, Tsunoda F, Tanaka N, Shiozawa T (2023) PPARα activator irbesartan suppresses the proliferation of endometrial carcinoma cells via SREBP1 and ARID1A. Oncol Res 31:239–253

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Zhou T, Xie Y, Hou X, Bai W, Li X, Liu Z, Man Q, Sun J, Fu D, Yan J, Zhang Z, Wang Y, Wang H, Jiang W, Gao S, Zhao T, Chang A, Wang X, Sun H, Zhang X, Yang S, Huang C, Hao J, Liu J (2023) Irbesartan overcomes gemcitabine resistance in pancreatic cancer by suppressing stemness and iron metabolism via inhibition of the Hippo/YAP1/c-Jun axis. J Exp Clin Cancer Res 42:111

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Titmuss E, Milne K, Jones MR, Ng T, Topham JT, Brown SD, Schaeffer DF, Kalloger S, Wilson D, Corbett RD, Williamson LM, Mungall K, Mungall AJ, Holt RA, Nelson BH, Jones SJM, Laskin J, Lim HJ, Marra MA (2023) Immune activation following Irbesartan treatment in a colorectal cancer patient: a case study. Int J Mol Sci 24:5869

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Gu L, Zhu Y, Lee M, Nguyen A, Ryujin NT, Huang JY, Pandit SK, Chamseddine S, Xiao L, Mohamed YI, Kaseb AO, Karin M, Shalapour S (2023) Angiotensin II receptor inhibition ameliorates liver fibrosis and enhances hepatocellular carcinoma infiltration by effector T cells. Proc Natl Acad Sci USA 120:e2300706120

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Gao Y, Wang H, Shen L, Xu H, Deng M, Cheng M, Wang J (2022) Discovery of benzo[d]isothiazole derivatives as novel scaffold inhibitors targeting the programmed cell death-1/programmed cell death-ligand 1 (PD-1/PD-L1) interaction through “ring fusion” strategy. Bioorg Chem 123:105769

    Article  CAS  PubMed  Google Scholar 

  86. Thuru X, Magnez R, Vergoten G, Bailly C (2023) A potential off-target effect of the Wnt/β-catenin inhibitor KYA1797K: PD-L1 binding and checkpoint inhibition. Biomed Hub 8:1–9

    Article  PubMed  PubMed Central  Google Scholar 

  87. Chopra C, Yodun T, Singh H, Singh B, Singh SK, Goutam U (2023) Raloxifene, a SERM targets PD-L1: an in-silico study. Am J Transl Res 15:5206–5215

    PubMed  PubMed Central  Google Scholar 

  88. Sobral PS, Luz VCC, Almeida JMGCF, Videira PA, Pereira F (2023) Computational approaches drive developments in immune-oncology therapies for PD-1/PD-L1 immune checkpoint inhibitors. Int J Mol Sci 24:5908

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Bianconi E, Riccio A, Ruta L, Bigiotti C, Carotti A, Moretti S, Cerra B, Gioiello A, Ferlin S, Puxeddu E, Macchiarulo A (2023) Turning a tumor microenvironment pitfall into opportunity: discovery of benzamidoxime as PD-L1 ligand with pH-dependent potency. Int J Mol Sci 24:5535

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

GV: Investigation; Visualization; Software. CB: Conceptualization; Visualization; Writing—original draft; Writing—review and editing.

Corresponding author

Correspondence to Christian Bailly.

Ethics declarations

Ethics approval and consent to participate

Not concerned.

Consent for publication

The two authors have approved the manuscript.

Competing interests

The authors declare no conflict of interest associated with this publication.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Fig. S1

. Binding map contacts for the different drugs and compounds bound to PD-L1 dimer. Fig. S2. Binding map contacts for the two best binding poses determined with Olmesartan. (left) The best binding pose, as shown in Fig. 4 and (right) the second-best binding pose.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vergoten, G., Bailly, C. Interaction of telmisartan and related sartans with the programmed cell death-ligand 1 (PD-L1) protein dimer: a molecular docking analysis. Futur J Pharm Sci 9, 118 (2023). https://doi.org/10.1186/s43094-023-00574-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43094-023-00574-1

Keywords