New horizons in cancer immunotherapy: The evolving role of R848 and R837 (Review)
- Authors:
- Published online on: October 31, 2024 https://doi.org/10.3892/mco.2024.2799
- Article Number: 4
-
Copyright: © Bhaliya et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 4.0].
Abstract
1. Introduction
Immunotherapy utilizes the body's immune system to act as a natural defense against cancer cells, which is notably different to traditional cancer treatments, including chemotherapy and radiotherapy. Advancements in immunotherapy have revolutionized disease management and cancer immunology. Results of clinical trials demonstrated that several types of immunotherapies, such as adoptive cellular therapy and immune checkpoint inhibitors (ICIs), exhibit potential in the treatment of cancer; however, efficacy may vary between patients and benefits may only be observed in certain groups (1).
Receptors, such as Toll-like receptors (TLRs), bridge the gap between innate and adaptive immunity, highlighting them as potential targets for therapeutic interventions. TLRs contribute to the first line of defense against pathogens, triggering signaling pathways that initiate immune and inflammatory responses (2).
The limited immunogenicity of cancer cells in combination with an immunosuppressive microenvironment is a pivotal protective factor that allows evasion of immune surveillance. Immune cells do not distinctly recognize tumor cells, and the tumor microenvironment (TME) notably hinders the infiltration or survival of immune cells (3).
TLRs are evolutionarily-conserved receptors that play a vital role in immune responses, particularly in recognizing microbes. TLRs belong to the family of pattern recognition receptors and are directly involved in regulating inflammatory responses, as well as activating innate or adaptive immune responses to eliminate infectious microorganisms and debris derived from cancer cells.
TLRs expressed on the cell membrane form an effective group with those located on the endosomal plasma membrane. These include TLR1, TLR2, TLR4, TLR5, TLR6, TLR10, TLR3, TLR7, TLR8 and TLR9. In cancer immunotherapy, the modulation of TLRs is employed to enhance the immune response against cancer cells. Thus, specific drugs or agents are designed to activate specific TLRs, thereby initiating an immune response that targets cancer. This strategy is commonly referred to as TLR agonism.
2. An overview of R848 and R837
R848 and R837 are synthetic compounds that modulate the immune response against cancer cells, acting as agonists for TLRs. R848, also known as Resiquimod, activates TLR7/8 receptors in various immune cells. R848 induces the anti-tumor response; thus, exhibiting potential as an immunotherapeutic agent. Notably, dendritic cells are actively involved in initiating and regulating immune responses, and these are stimulated by R848. However, immunotherapies are only effective in a specific TME, and R848 remodels the TME to aid in immunotherapy (4,5).
R837, also known as Imiquimod, also activates TLR7/8 receptors in various immune cells. R837 is well-established as a combination therapy with chemotherapy and photochemical therapy, and is used to improve immunotherapeutic efficacy (6).
3. Mechanisms of action
R848 and R837 are imidazoquinolines that act as agonists for TLR7 or TLR8. Their drug-receptor interaction activates immune cells and creates an environment that stimulates T helper cell (Th1) immune responses. In humans, R848 functions as an agonist for both TLR7 and TLR8; however, in mice, it acts as a preferential agonist to TLR7. R837 is a selective antagonist of TLR7 only. The Myd88-dependent signaling pathway is utilized by R848 and R837 to activate TLR7/TLR8, which in turn activates transcription factors; namely, NF-κB and JNK. Ultimately, this pathway leads to the production of Th1 cytokines, such as type I interferon (IFN I), interleukin (IL)-6, IL-12, IFN-γ and tumor necrosis factor-α (TNF-α), while suppressing the expression and secretion of Th2 cytokine, IL-4. Moreover, Th1 cytokines trigger the release of several cytokines that facilitate innate and acquired immunity, through activation of monocytes, macrophages, and plasmacytoid dendritic cells via TLR7/8. In addition, the activation of TLRs stimulates the activity of antigen-presenting cells (APCs). APCs present antigens to T-cells, thereby initiating an immune response. This, in turn, triggers tumor-associated macrophages (TAMs) that modulate the TME through activating innate immune responses. Thus, tumor growth is inhibited and the TME is disrupted via R848- and R837-mediated activation of TAMs (Fig. 1) (7).
4. Preclinical studies in cancer immunotherapy using R848 and R837
Several preclinical studies have previously been conducted to study the role and effects of R848 and R837 in cancer immunotherapy. Ye et al (8) evaluated the impact of intravenous R848 and stereotactic body radiation therapy (SBRT) on tumor growth and immune response in primary pancreatic tumors, using murine models of orthotopic pancreatic ductal adenocarcinoma (PDAC). Results of this previous study demonstrated that the combination of R848 and SBRT significantly activated the pancreatic TME, increased levels of IFN-γ, granzyme B and chemokine ligand 5, and decreased levels of IL-4, IL-6 and IL-10. In addition, R848 and SBRT increased tumor antigen-specific CD8+ T-cells, reduced the number of regulatory T-cells and improved antigen-presenting cell maturation (8). Moreover, Mottas et al (9) utilized amphiphilic polymer and monoblock peptide nanoparticles to encapsulate and deliver R848 and R837 to human and murine cancer cell lines. Results of this previous study revealed that R848 and R837 targeted blood-borne macrophages and promoted tumor regression in experimental gliomas without adaptive immunity. In a further study, nanoparticles consisting of polyglycolic acid (PLGA) combined with immunostimulant granulocyte-colony stimulating factor (ICG) were used as a carrier for R848 via near-infrared (NIR). In both human and murine cell lines for prostate cancer, a favorable immunohistochemical response was observed. The combination of nanoparticles containing cancer cell membranes derived from surgically excised cancer cells and mesoporous polydopamine carrying R848 were also studied, and the results revealed that this nano vaccine exhibited potent phagocytosis, isotype targeting and tumor-specific immune responses when exposed to NIR. In addition, this combination promoted photothermal immunotherapy at the tumor site (10). Results of a further previous study revealed that R837 in combination with hematoporphyrin monomethyl ether arrested primary tumor progression and prevented lung metastasis, when administered via biomedical ultrasound (7,11-13). Thus, the results of preclinical studies have led to the development of clinical trials that aimed to determine the efficacy and safety of R837 and R848 in healthy human volunteers.
5. Clinical trials
R837 and R848, initially explored for their potential in the treatment of herpes simplex virus (HSV), exhibited limitations in clinical trials due to insufficient efficacy and notable side effects. Despite evidence suggesting success in the treatment of acute and chronic HSV lesions, off-label usage became common (14-16). For example, R837 demonstrated potential in the treatment of vaginal or cervical human papillomavirus (HPV) lesions, and the effects were more apparent in combination with photodynamic therapy or HPV therapeutic vaccines for HPV-16-mediated vulvar intraepithelial neoplasia (17). However, significant side effects, such as severe vulval erythema, and unsuccessful trials in patients with cervical intraepithelial neoplasia were reported (18).
In a clinical trial, R837 demonstrated disease clearance rates exceeding 50%, highlighted by increased levels of immune cells in biopsied lesions. Approval from the Food and Drug Administration (FDA) was granted for the treatment of basal cell carcinoma in 2004, with high disease clearance rates observed in Phase III clinical trials (19). Notably, Th1 polarization, enhanced IFN I signaling and immune cell recruitment were observed in skin biopsies. R848 also demonstrated high levels of efficacy in cutaneous T-cell lymphoma treatment, inducing significant improvements in patients (20). Notably, clinical trials that aim to explore novel combinations and delivery strategies of R837 and R848 are ongoing (Table I).
Table IRegistered clinical trials that aimed to determine the safety and efficacy of Imiquimod and Resiquimod in the treatment of various cancers. |
The SINS study (trial no. NCT00066872) was a multi-centric, randomized study including 500 patients with basal skin cell cancer, who participated for 18 months. Patients were categorized depending on the presence of superficial or nodular lesions and the participating center. Patients were randomly assigned to two treatment arms, and those with superficial lesions were treated for 6 weeks, while patients with nodular lesions were treated for 12 weeks. Patients in treatment arm one received topical Imiquimod, and this was applied to a single lesion once daily. Surgical excision was offered to patients who exhibited recurrence or failed early treatment. Patients in treatment arm two underwent surgical excision. Follow-up was carried out at 6, 12 and 18 weeks, every 6 months for two years, and every 5 years. Results of this previous study revealed that Imiquimod was effective in ~90% of patients with large superficial basal cell carcinoma (diameter, >4 cm). Notably, Imiquimod was considered the preferred course of treatment; however, it was only marginally less effective than excisional surgery due to the associated cosmetic outcomes (21).
Females who are yet to reach menopause are primarily affected by vulval intraepithelial neoplasia, a pre-malignant disease. This condition is symptomatic and invasive, leading to challenges in clinical practice. Treatment goals include symptom relief and inhibition of disease spread. Surgery is frequently used to cure this malignant condition; however, it may be deformative and exhibits a high recurrence rate. Recent pilot and small-scale studies using novel topical therapies with Imiquimod as a non-surgical alternative have demonstrated positive outcomes.
A randomized Phase II multi-centric clinical trial (trial no. ISRCTN34420460) included 204 patients. Patients received topical treatment of Imiquimod or Cidofovir for up to 24 weeks. Every 6 weeks, patients were reviewed and evaluated, and treatment was administered for up to 24 weeks if there was no full response. Notably, a full response was established histologically at 30 weeks following therapy initiation. In total, 156 patients (87%) followed the treatment plan for >6 weeks (78 in each group). Prior to the initial 6-week safety evaluation, 5 patients in the Imiquimod group and 7 patients in the Cidofovir group stopped responding to follow-up or withdrew from the study. Of the 84 patients assigned to the Cidofovir treatment group, 31 (37%) experienced adverse events of grade 3 or above, whereas 39 (46%) of the 84 patients assigned to the Imiquimod treatment group experienced adverse events of grade 3 or above. Notably, the most common grade 3 and 4 events were headache, pruritus, pain in the vulva and exhaustion. Further investigations into the use of Cidofovir and Imiquimod are required, as these reagents demonstrate high levels of efficacy and safety, and may exhibit potential as an alternative to surgical intervention in the treatment of vulval intraepithelial neoplasia (22).
An open, blinded, non-controlled clinical trial (trial no. NL-OMON3398) aimed to determine whether Imiquimod, granulocyte-macrophage colony-stimulating factor, IL-2 and IFN-α induce peritumoral responses when used in conjunction with a peptide vaccination. The type of human leukocyte antigen and gene expression of the tumor determined whether the MAGE-3.A1 peptide, NA17.A2 peptide or both peptide vaccines would be used. If both antigens were expressed, the patient was treated with both peptide vaccines. This combination triggers cytolytic T-cell (CTL) responses, which were evaluated through comparing the frequency of either anti-MAGE-3. A1 or anti-NA17.A2 CTLp in the pre- and post-immune blood of patients. This trial exhibited limitations, as there were a small number of subjects, and technical problems led to unreliable data. Thus, the trial was subsequently terminated (23).
NCT03196180 was an early Phase I, open-label clinical trial involving 13 patients with high-grade cervical intraepithelial neoplasia, who were treated with topical fluorouracil and Imiquimod. In patients with precancerous cervical lesions, topical fluorouracil and Imiquimod ointment may lead to positive outcomes due to fewer associated adverse effects. For the treatment of high-grade cervical squamous intraepithelial lesions, a once-weekly intravaginal application of 5-fluorouracil was used, and this was alternated with a once-weekly application of Imiquimod. For 8-16 weeks, 5-fluorouracil and Imiquimod treatments were alternated. The median age of participants was 27±2.9 years. There were no grade 3/4 adverse events, six (46.15%) grade 2 adverse events and all individuals reported grade 1 adverse events. In total, 3 patients (23.08%) reported specific adverse events. Notably, 10 patients (90.91%) exhibited histologic regression to cervical intraepithelial neoplasia (CIN), and 7 patients (63.63%) tested negative for hr-HPV following the study. Further investigations are required for the use of topical treatments in CIN 2/3 as alternatives or adjuncts to surgical therapy (24).
The NCT01676831 Phase I clinical trial was an open-label, dose-ranging trial including patients with early-stage (IA-IIA) cutaneous T-cell lymphoma (CTCL). This study investigated the safety and efficacy of Resiquimod gel at concentrations of 0.06 and 0.03%, administered to individuals with early-stage cutaneous T-cell lymphoma lesions. In 75% of patients, considerable improvements were observed in treated lesions, and 30% of patients experienced total clearance of lesions. In addition, Resiquimod also induced the regression of untreated lesions. According to the modified Severity-Weighted Assessment Tool, 92% of patients exhibited a ≥50% improvement in body surface area involved, and 2 patients experienced full clearance of the disease. In total, 4 out of 5 patients with follicotropic disease demonstrated improvements. In addition, 90% of patients exhibited reduced clonal malignant T-cells, and 30% exhibited completely eradicated malignant T-cells, according to T-cell receptor sequencing and flow cytometry analysis of treated lesions. Elevated responses were associated with enhanced cutaneous T-cell effector functions, enhanced natural killer cell functions, and the recruitment and growth of benign T-cell clones in treated skin. Cytokines were also associated with persistent clinical inflammation in patients with malignant T-cells that had been almost completely eliminated. A systemic response to medication was associated with the enhanced activation of circulating dendritic cells in 50% of patients. Further Phase II trials using topical Resiquimod in early-stage CTCL are required (23). Diseases that may be targeted by R837 and R848 are summarized in Fig. 2.
6. Combination therapy as an advanced therapeutic approach
ICIs
Previous research has focused on the combination of the TLR7/8 agonist, R848, with ICIs in cancer cells. Hänel et al (24) demonstrated that the combination of R848 with poly(I: C) was more effective in eliciting an immunostimulatory response than when R848 was used alone. This combination induces the reprogramming of macrophages into M1 hot-antitumor effectors through signal transducer and activator of transcription 1 activation (9). This study included patients with lung cancer resistant to immune checkpoint blockade (ICB) (25).
TLRs
In a preclinical study using a humanized mouse model, Poly(I: C) combined with R848 increased the expression of various cytokines, compared with Poly(I: C) or R848 administered alone (26). In addition, the synergistic use of Poly(I: C) and R848 with E7 DNA promoted the regression of tumors, and increased the levels of antigen-specific IFN-γ and non-specific intertumoral IL-12(27). Results of a previous study also demonstrated that the combination of TLR agonists with prostaglandins and TLR3 Poly(I: C)/R848/PGE2 aided dendritic cells in the mobilization and secretion of cytokines, including IL-12 (p70) (28).
In a murine model, treatment with biocompatible anchors for the membrane (BAM), R848, Poly(I: C) and lipoteichoic acid led to an 83% eradication of advanced progressive melanoma and reduced tumor recurrence, when in contact with the surface of tumor cells (29).
Chemotherapy and radiotherapy
TLR7/8 agonist, R848, plays a crucial role in the action of oxaliplatin. Oxaliplatin is a chemotherapeutic drug that promotes the maturation of myeloid-derived suppressor cells (MDSCs) into M1-like macrophages, which play key roles in various immune reactions (30). A combination of Resiquimod with chemotherapeutic agents exhibits potential in the treatment of cancer, as dosage can be altered depending on patient outcomes. At present, research is focused on the use of chemotherapeutic drugs in combination with TLR-mediated immunotherapy (31). Notably, reduced tumor growth was observed following treatment with R837 in combination with γ-ionizing radiation (IR). This combination of treatment increased autophagy and the number of CD8+ T-cells, and decreased Tregs and MDSCs (31).
Adjuvant therapy
In the presence of R837-containing nanoparticles as adjuvants, PLGA-ICG-R837 nanoparticles function as vaccines. PLGA-ICG-R837 nanoparticles may trigger NIR laser-induced photothermal ablation of primary tumors and tumor-associated antigens. In mice, PLGA-ICG-R837 nanoparticles target tumor cells; thus, limiting metastasis. This strategy led to the establishment of a robust immune memory effect, potentially protecting against tumor recurrence following initial elimination (32).
Dual-functional nanoparticles combining partial thromboplastin time and immunotherapy components (PLGA-ICG-R848 NPs) promoted the maturation of dendritic cells, as evidenced by increased proportions of CD11c+CD86+ and CD11c+CD80+ cells. This strategy exhibits potential for improving the immune response against tumor cells (33).
TLR7/8 agonist-containing nanoparticles have also been developed for systemic administration. Results of a previous study described semiconducting polymer nano adjuvant (SPNIIR) for second near-infrared (NIR-II) photothermal immunotherapy. When exposed to NIR-II light, SPNIIR generated heat to erode tumors, induced immunogenic cell death and released R848, enhancing the maturation of dendritic cells. This approach effectively inhibited the growth of primary and distant tumors, and eliminated lung metastasis in a murine tumor model (34).
Zhang et al (35) explored intravenous TLR7/8 agonist administration using thermosensitive liposomes (TSLs) to enhance CD8+ T-cell infiltration. Mice treated with R848-loaded TSLs combined with local hyperthermia and αPD-1 exhibited a 3-fold increase in median survival, compared with controls. Mice treated with R848-TSLs and αPD-1 developed immunity to NDL cells, and remained tumor-free when NDL tumors were reintroduced. Results of a previous study demonstrated that polydopamine-based PDA-PEG-R848-CD nanoparticles impacted 4T1 breast tumors under NIR laser irradiation, generated tumor-associated antigens and significantly improved the efficacy of PD-L1 checkpoint blockade therapy, activating innate and adaptive immune systems in vivo (36,37).
7. Limitations of R837 and R848
Applications of R837 and R848 may be limited due to low levels of efficacy and associated levels of toxicity in cells. R837 and R848 may exhibit potential in promoting immune responses; however, this may lead to the development of specific antibodies. Notably, this process is known as an antibody-drug reaction (ADR). ADRs may reduce the efficacy of the drug and cause undesirable adverse effects. Thus, to moderate the risk of ADRs, further in vitro investigations are required to establish and eliminate immunogenic epitopes (38).
Results of previous studies have demonstrated the potential of these drugs as cancer therapies. However, systemic administration of these TLR7/8 agonists remains challenging. Although TLR7 tolerance has been previously demonstrated, the tolerance induction mechanism remains unclear. Thus, further investigations are required to determine the efficacy of R837 and R848 in cancer. In addition, the association between TLR expression, agonist activity and circadian rhythm are yet to be explored in the context of TLR7/8 agonism.
TLRs play a complex, multifaceted role in the TME. Immune cell-expressed TLRs may support immunosurveillance through promoting and activating both innate and adaptive immune effectors. However, myeloid-derived suppressor cells are recruited by TLR7 expressed by malignant cells; thus, promoting the growth and spread of tumors. The pharmacokinetic characteristics of the small-molecule adjuvants cause substantial systemic inflammation, which may impact their use in clinical practice. However, results of a previous study revealed that the covalent attachment of TLR7/8 agonists to polymer scaffolds may modify the associated safety profile and increase levels of clearance. When encapsulated in nanoparticles, R837 promoted the development of DCs. However, this previous study did not investigate drug reactions or ADRs (39,40).
Moreover, TLRs produced by tumor cells may interact with stimuli to promote the growth of tumors. Imidazoquinolines-based small molecules stimulate immune responses; however, their pharmacokinetic profile leads to systemic inflammation, ultimately impacting their use in clinical practice. Tumor immunotherapy exhibits limitations in practice due to the absence of biomarkers. Existing biomarkers, such as microsatellite instability, PD-L1, genetic mutations, and tumor mutational burden also exhibit limitations, and these are impacted by the type of cancer and cancer heterogeneity (41). The aforementioned biomarkers may not be present in all patients; thus, further investigations are required to establish reliable biomarkers of tumor immunotherapy (42). Integrative biomarker frameworks consolidating safety profiling, clinical history and prescient calculations may aid in the development of effective treatment options (10).
Patient selection and stratification are crucial in clinical trials. Notably, patients may be grouped according to age, cancer type and stage. Clinical trials including patients that are well-suited to treatments may lead to the development of customized treatment plans, and further demonstrate the integrity of the trial (43).
Clinical trials involving R848 and R837 should focus on stratifying patients according to cancer type, cancer stage, patient well-being and the presence of any comorbid conditions (44). Patients may also be stratified according to the presence of specific biomarkers, medical history or previous adverse events that may impact treatment outcomes (45).
8. Future directions of the R848 and R837 immunotherapy strategy
Drug delivery systems, such as nanoparticles or liposomes, may be optimized to enhance the targeted delivery of R848 and R837, and to control off-target effects. Administering a sustainable release system of drugs may improve therapeutic outcomes via the reduction in the frequency of treatments and improved convenience for patients. In addition, these factors may enhance compliance within immunotherapeutic practice. Developing systems that are FDA-approved will also lead to increased uptake in clinical practice. At present, systems that use FDA-approved lipids to deliver mRNA to dendritic cells are ongoing in clinical trials (46,47). Immunotherapies specific to patient profiles may be managed through biomarker-driven approaches. These approaches include genetic profiling and immune system analysis. In addition, personalized cancer vaccines may be established via neoantigen identification advancements. Numerous immunotherapeutic approaches, such as ICIs, agonistic antibodies, cancer vaccines, lymphocyte-activating cytokines, CAR T-cells and bispecific antibodies exhibit potential in the treatment of cancer (48). R848 and R837 may also exhibit potential as an immunotherapy, with precise specifications and a targeted approach. Moreover, the combination of R848 and R837 with chemotherapy, radiation therapy, nanoparticle-mediated immunoadjuvant therapy or phototherapy may exhibit potential in the treatment of cancer (6,49).
In the preliminary stage of malignancy, the TME is more susceptible to treatment, and cancer cells may not develop immunosuppressive mechanisms. Thus, introducing agonists in the early stages of disease may lead to earlier activation of immune cells, such as dendritic cells, and the initiation of an adaptive immune response. Notably, tumors may be recognized by immune cells during remission, demonstrating the potential of TLR agonists in the early stages of disease (50,51).
9. Conclusions
Numerous previous studies have focused on TLR7/8 signaling in immune cells and progenitor cells, and demonstrated that agonists exhibit high levels of efficacy in tumor cells, in which TLR7/8 signaling either promotes or inhibits tumor growth.
R848 and R837 are considered innate immunity activators that target tumor growth and metastases, and these exhibit potential as a novel therapeutic option for the treatment of cancer. Clinical trials focused on the safety and efficacy of these molecules are ongoing. Notably, Imiquimod and Resiquimod exhibit increased levels of anti-tumor activity when used in combination therapies; thus, R848 and R837 may exhibit improved outcomes when used in combination. Therapeutic drug delivery using nanoparticles may be limited due to the off-target effects of R848 and R837, and improvements in targeted delivery and control are required. In conclusion, further investigations into drug delivery, personalized approaches and tumor resistance are required for the development of R848 and R837 as an immunotherapy.
Acknowledgements
Not applicable.
Funding
Funding: No funding was received.
Availability of data and materials
Not applicable.
Authors' contributions
KB was responsible for study design and writing the manuscript. MA was responsible for editing the manuscript. MW and AM was responsible for final revision of the manuscript. Data authentication is not applicable. All authors read and approved the final version of the manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Zhang Y and Zhang Z: The history and advances in cancer immunotherapy: Understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell Mol Immunol. 17:807–821. 2020.PubMed/NCBI View Article : Google Scholar | |
Pahlavanneshan S, Sayadmanesh A, Ebrahimiyan H and Basiri M: Toll-like receptor-based strategies for cancer immunotherapy. J Immunol Res. 2021(9912188)2021.PubMed/NCBI View Article : Google Scholar | |
Chen X, Zhang Y and Fu Y: The critical role of Toll-like receptor-mediated signaling in cancer immunotherapy. Med Drug Disc. 14(100122)2022. | |
Spyvee M, Hawkins LD and Ishizaka ST: Chapter 12 - Modulators of Toll-Like Receptor (TLR) Signaling. In: Annual Reports in Medicinal Chemistry. Vol 45. Academic Press, pp191-207, 2010. | |
Ryu KA, Stutts L, Tom JK, Mancini RJ and Esser-Kahn AP: Stimulation of innate immune cells by light-activated TLR7/8 agonists. J Am Chem Soc. 136:10823–10825. 2014.PubMed/NCBI View Article : Google Scholar | |
Bhagchandani S, Johnson JA and Irvine DJ: Evolution of Toll-like receptor 7/8 agonist therapeutics and their delivery approaches: From antiviral formulations to vaccine adjuvants. Adv Drug Deliv Rev. 175(113803)2021.PubMed/NCBI View Article : Google Scholar | |
Tie Y, Tang F, Wei YQ and Wei XW: Immunosuppressive cells in cancer: Mechanisms and potential therapeutic targets. J Hematol Oncol. 15(61)2022.PubMed/NCBI View Article : Google Scholar | |
Ye J, Mills BN, Qin SS, Garrett-Larsen J, Murphy JD, Uccello TP, Han BJ, Vrooman TG, Johnston CJ, Lord EM, Belt BA, et al: Toll-like receptor 7/8 agonist R848 alters the immune tumor microenvironment and enhances SBRT-induced antitumor efficacy in murine models of pancreatic cancer. J Immunother Cancer. 10(e004784)2022.PubMed/NCBI View Article : Google Scholar | |
Mottas I, Bekdemir A, Cereghetti A, Spagnuolo L, Yang YS, Müller M, Irvine DJ, Stellacci F and Bourquin C: Amphiphilic nanoparticle delivery enhances the anticancer efficacy of a TLR7 ligand via local immune activation. Biomaterials. 190:111–120. 2019.PubMed/NCBI View Article : Google Scholar | |
Wang DR, Wu XL and Sun YL: Therapeutic targets and biomarkers of tumor immunotherapy: Response versus non-response. Sig Transduct Target Ther. 7(331)2022.PubMed/NCBI View Article : Google Scholar | |
Leśniak M, Lipniarska J, Majka P, Kopyt W, Lejman M and Zawitkowska J: The Role of TRL7/8 agonists in cancer therapy, with special emphasis on hematologic malignancies. Vaccines (Basel). 11(277)2023.PubMed/NCBI View Article : Google Scholar | |
Sun H, Li Y, Zhang P, Xing H, Zhao S, Song Y, Wan D and Yu J: Targeting toll-like receptor 7/8 for immunotherapy: Recent advances and prospectives. Biomark Res. 10(89)2022.PubMed/NCBI View Article : Google Scholar | |
Karnik I, Her Z, Neo SH, Liu WN and Chen Q: Emerging preclinical applications of humanized mouse models in the discovery and validation of novel immunotherapeutics and their mechanisms of action for improved cancer treatment. Pharmaceutics. 15(1600)2023.PubMed/NCBI View Article : Google Scholar | |
Deza G, Martin-Ezquerra G, Curto-Barredo L, García JV and Pujol RM: Successful treatment of hypertrophic herpes simplex genitalis in an HIV-infected patient with topical Imiquimod. J Dermatol. 42:1176–1178. 2015.PubMed/NCBI View Article : Google Scholar | |
Miller RL, Imbertson LM, Reiter MJ and Gerster JF: Treatment of primary herpes simplex virus infection in guinea pigs by imiquimod. Antiviral Res. 44:31–42. 1999.PubMed/NCBI View Article : Google Scholar | |
Schacker TW, Conant M, Thoming C, Stanczak T, Wang Z and Smith M: Imiquimod 5-Percent cream does not alter the natural history of recurrent herpes genitalis: A phase II, Randomized, double-blind, placebo-controlled study. Antimicrob Agents Chemother. 46:3243–3248. 2002.PubMed/NCBI View Article : Google Scholar | |
Winters U, Daayana S, Lear JT, Tomlinson AE, Elkord E, Stern PL and Kitchener HC: Clinical and immunologic results of a phase II trial of sequential imiquimod and photodynamic therapy for vulval intraepithelial neoplasia. Clin Cancer Res. 14:5292–5299. 2008.PubMed/NCBI View Article : Google Scholar | |
Wouters T, Hendriks N, Koeneman M, Kruse AJ, van de Sande A, van Beekhuizen HJ, Gerestein KG, Bekkers RLM and Piek JMJ: Systemic adverse events in Imiquimod use for cervical intraepithelial neoplasia-A case series. Case Rep Womens Health. 21(e00105)2019.PubMed/NCBI View Article : Google Scholar | |
Lebwohl M, Dinehart S, Whiting D, Lee PK, Tawfik N, Jorizzo J, Lee JH and Fox TL: Imiquimod 5% cream for the treatment of actinic keratosis: Results from two phase III, randomized, double-blind, parallel group, vehicle-controlled trials. J Am Acad Dermatol. 50:714–721. 2004.PubMed/NCBI View Article : Google Scholar | |
Geisse J, Caro I, Lindholm J, Golitz L, Stampone P and Owens M: Imiquimod 5% cream for the treatment of superficial basal cell carcinoma: Results from two phase III, randomized, vehicle-controlled studies. J Am Acad Dermatol. 50:722–733. 2004.PubMed/NCBI View Article : Google Scholar | |
Ackerman SE, Gonzalez JC, Gregorio JD, Paik JC, Hartmann FJ, Kenkel JA, Lee A, Luo A, Pearson CI, Nguyen ML, et al: Abstract 1559: TLR7/8 immune-stimulating antibody conjugates elicit robust myeloid activation leading to enhanced effector function and anti-tumorimmunity in pre-clinical models. Cancer Res. 79 (13 Suppl)(S1559)2019. | |
Ackerman SE, Pearson CI, Gregorio JD, Gonzalez JC, Kenkel JA, Hartmann FJ, Luo A, Ho PY, LeBlanc H, Blum LK, et al: Immune-stimulating antibody conjugates elicit robust myeloid activation and durable antitumor immunity. Nat Cancer. 2:18–33. 2021.PubMed/NCBI View Article : Google Scholar | |
Dudek AZ, Yunis C, Harrison LI, Kumar S, Hawkinson R, Cooley S, Vasilakos JP, Gorski KS and Miller JS: First in human phase I trial of 852A, a novel systemic toll-like receptor 7 agonist, to activate innate immune responses in patients with advanced cancer. Clin Cancer Res. 13:7119–7125. 2007.PubMed/NCBI View Article : Google Scholar | |
Hänel G, Angerer C, Petry K, Lichtenegger FS and Subklewe M: Blood DCs activated with R848 and poly (I: C) induce antigen-specific immune responses against viral and tumor-associated antigens. Cancer Immunol Immunother. 71:1705–1718. 2022.PubMed/NCBI View Article : Google Scholar | |
Anfray C, Mainini F, Digifico E, Maeda A, Sironi M, Erreni M, Anselmo A, Ummarino A, Gandoy S, Expósito F, et al: Intratumoral combination therapy with poly(I:C) and Resiquimod synergistically triggers tumor-associated macrophages for effective systemic antitumoral immunity. J Immunother Cancer. 9(e002408)2021.PubMed/NCBI View Article : Google Scholar | |
Pearson FE, Chang K, Minoda Y, Rojas IML, Haigh OL, Daraj G, Tullett KM and Radford KJ: Activation of human CD141(+) and CD1c(+) dendritic cells in vivo with combined TLR3 and TLR7/8 ligation. Immunol Cell Biol. 96:390–400. 2018.PubMed/NCBI View Article : Google Scholar | |
Sajadian A, Tabarraei A, Soleimanjahi H, Fotouhi F, Gorji A and Ghaemi A: Comparing the effect of Toll-like receptor agonist adjuvants on the efficiency of a DNA vaccine. Arch Virol. 159:1951–1960. 2014.PubMed/NCBI View Article : Google Scholar | |
Gierlich P, Lex V, Technau A, Keupp A, Morper L, Glunz A, Sennholz H, Rachor J, Sauer S, Marcu A, et al: Prostaglandin E2 in a TLR3- and 7/8-agonist-based DC maturation cocktail generates mature, cytokine-producing, migratory DCs but impairs antigen cross-presentation to CD8(+) T-cells. Cancer Immunol Immunother. 69:1029–1042. 2020.PubMed/NCBI View Article : Google Scholar | |
Caisova V, Uher O, Nedbalova P, Jochmanova I, Kvardova K, Masakova K, Krejcova G, Padoukova L, Chmelar J, Kopecky J and Ženka J: Effective cancer immunotherapy based on the combination of TLR agonists with stimulation of phagocytosis. Int Immunopharmacol. 59:86–96. 2018.PubMed/NCBI View Article : Google Scholar | |
Liu Z, Xie Y, Xiong Y, Liu S, Qiu C, Zhu Z, Mao H, Yu M and Wang X: TLR 7/8 agonist reverses oxaliplatin resistance in colorectal cancer via directing the myeloid-derived suppressor cells to tumoricidal M1-macrophages. Cancer Lett. 469:173–185. 2020.PubMed/NCBI View Article : Google Scholar | |
Liu C, Han C and Liu J: The role of toll-like receptors in oncotherapy. Oncol Res. 27:965–978. 2019.PubMed/NCBI View Article : Google Scholar | |
Cho JH, Lee HJ, Ko HJ, Yoon BI, Choe J, Kim KC, Hahn TW, Han JA, Choi SS, Jung YM, et al: The TLR7 agonist Imiquimod induces anti-cancer effects via autophagic cell death and enhances anti-tumoral and systemic immunity during radiotherapy for melanoma. Oncotarget. 8:24932–24948. 2017.PubMed/NCBI View Article : Google Scholar | |
Chen Q, Xu L, Liang C, Wang C, Peng R and Liu Z: Photothermal therapy with immune-adjuvant nanoparticles together with checkpoint blockade for effective cancer immunotherapy. Nat Commun. 7(13193)2016.PubMed/NCBI View Article : Google Scholar | |
Lin W, Li C, Xu N, Watanabe M, Xue R, Xu A, Araki M, Sun R, Liu C, Nasu Y and Huang P: Dual-Functional PLGA nanoparticles co-loaded with indocyanine green and resiquimod for prostate cancer treatment. Int J Nanomed. 16:2775–2787. 2021.PubMed/NCBI View Article : Google Scholar | |
Zhang H, Tang WL, Kheirolomoom A, Fite BZ, Wu B, Lau K, Baikoghli M, Raie MN, Tumbale SK, Foiret J, et al: Development of thermosensitive resiquimod-loaded liposomes for enhanced cancer immunotherapy. J Control Release. 330:1080–1094. 2021.PubMed/NCBI View Article : Google Scholar | |
Li J, Yu X, Jiang Y, He S, Zhang Y, Luo Y and Pu K: Second near-infrared photothermal semiconducting polymer nanoadjuvant for enhanced cancer immunotherapy. Adv Mater. 33(e2003458)2021.PubMed/NCBI View Article : Google Scholar | |
Noman MZ, Parpal S, Van Moer K, Xiao ML, Yu Y, Viklund J, De Milito A, Hasmim M, Andersson M, Amaravadi RK, et al: Inhibition of Vps34 reprograms cold into hot inflamed tumors and improves anti-PD-1/PD-L1 immunotherapy. Sci Adv. 6(eaax7881)2020.PubMed/NCBI View Article : Google Scholar | |
Shakhnovich V, Meibohm B, Rosenberg A, Kierzek AM, Hasenkamp R, Funk RS, Thalhauser CJ, van der Graaf PH, Wang YC and Hamuro L: Immunogenicity in clinical practice and drug development: When is it significant? Clin Transl Sci. 13:219–223. 2020.PubMed/NCBI View Article : Google Scholar | |
Zhang R, Jia M, Lv H, Li M, Ding G, Cheng G and Li J: Assembling Au8 clusters on surfaces of bifunctional nanoimmunomodulators for synergistically enhanced low dose radiotherapy of metastatic tumor. J Nanobiotechnology. 22(20)2024.PubMed/NCBI View Article : Google Scholar | |
Zahm CD, Colluru VT, McIlwain SJ, Ong IM and McNeel DG: TLR stimulation during T-cell activation lowers PD-1 expression on CD8(+) T-cells. Cancer Immunol Res. 6:1364–1374. 2018.PubMed/NCBI View Article : Google Scholar | |
Costa Svedman F, Jalsenius M, Höiom V, Grozman V, Bergqvist M, Söderdahl F, Eriksson H, Rotstein S, Ny L, Ascierto PA, et al: Plasma thymidine kinase activity as a novel biomarker in metastatic melanoma patients treated with immune checkpoint inhibitors. Cancers (Basel). 14(702)2022.PubMed/NCBI View Article : Google Scholar | |
Rizzo A, Ricci AD and Brandi G: Pd-L1, TMB, MSI, and other predictors of response to immune checkpoint inhibitors in biliary tract cancer. Cancers (Basel). 13(553)2021.PubMed/NCBI View Article : Google Scholar | |
Shen X and Zhao B: Efficacy of PD-1 or PD-L1 inhibitors and PD-L1 expression status in cancer: Meta-analysis. BMJ. 362(k3529)2018.PubMed/NCBI View Article : Google Scholar | |
Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2021. CA Cancer J Clin. 71:7–33. 2021.PubMed/NCBI View Article : Google Scholar | |
Wang X, Piantadosi S, Le-Rademacher J and Mandrekar SJ: Statistical considerations for subgroup analyses. J Thorac Oncol. 16:375–380. 2021.PubMed/NCBI View Article : Google Scholar | |
Riley RS, June CH, Langer R and Mitchell MJ: Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov. 18:175–196. 2019.PubMed/NCBI View Article : Google Scholar | |
Jin SM, Lee SN, Kim JE, Yoo YJ, Song C, Shin HS, Phuengkham H, Lee CH, Um SH and Lim YT: Overcoming chemoimmunotherapy-induced immunosuppression by assemblable and depot forming immune modulating nanosuspension. Adv Sci (Weinh). 8(2102043)2021.PubMed/NCBI View Article : Google Scholar | |
Fang X, Lan H, Jin K, Gong D and Qian J: Nanovaccines for cancer prevention and immunotherapy: An update review. Cancers (Basel). 14(3842)2022.PubMed/NCBI View Article : Google Scholar | |
Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon RA, Reed K, et al: Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 369:122–133. 2013.PubMed/NCBI View Article : Google Scholar | |
Muraoka D, Seo N, Hayashi T, Tahara Y, Fujii K, Tawara I, Miyahara Y, Okamori K, Yagita H, Imoto S, et al: Antigen delivery targeted to tumor-associated macrophages overcomes tumor immune resistance. J Clin Invest. 129:1278–1294. 2019.PubMed/NCBI View Article : Google Scholar | |
Garrido-Martin EM, Mellows TWP, Clarke J, Ganesan AP, Wood O, Cazaly A, Seumois G, Chee SJ, Alzetani A, King EV, et al: M1hot tumor-associated macrophages boost tissue-resident memory T cells infiltration and survival in human lung cancer. J Immunother Cancer. 8(e000778)2020.PubMed/NCBI View Article : Google Scholar |