Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Immunotherapy utilizing checkpoint inhibitors has shown remarkable success in the treatment of cancers. In addition to immune checkpoint inhibitors, immune co-stimulation has the potential to enhance immune activation and destabilize the immunosuppressive tumor microenvironment. CD137, also known as 4-1BB, is one of the potent immune costimulatory receptors that could be targeted for effective immune co-stimulation. The interaction of the 4-1BB receptor with its natural ligand (4-1BBL) generates a strong costimulatory signal for T cell proliferation and survival. 4-1BBL lacks costimulatory activity in soluble form. To obtain co-stimulatory activity in soluble form, a recombinant 4-1BBL protein was generated by fusing the extracellular domains of murine 4-1BBL to a modified version of streptavidin (SA-4-1BBL). Treatment with SA-4-1BBL inhibited the development of lung tumors in A/J mice induced by weekly injections of the tobacco carcinogen NNK for eight weeks. The inhibition was dependent on the presence of T cells and NK cells; depletion of these cells diminished the SA-4-1BBL antitumor protective effect. The number of lung tumor nodules was significantly reduced by the administration of SA-4-1BBL to mice during ongoing exposure to NNK. The data presented in this paper suggest that utilizing an immune checkpoint stimulator as a single agent generate a protective immune response against lung cancer in the presence of a carcinogen. More broadly, this study suggests that immune checkpoint stimulation can be extended to a number of other cancer types, including breast and prostate cancers, for which improved diagnostics can detect disease at the preneoplastic stage.

Free full text 


Logo of ciiLink to Publisher's site
Cancer Immunol Immunother. 2023 Nov; 72(11): 3567–3579.
Published online 2023 Aug 22. https://doi.org/10.1007/s00262-023-03507-2
PMCID: PMC10991934
PMID: 37605009

A novel agonist of 4-1BB costimulatory receptor shows therapeutic efficacy against a tobacco carcinogen-induced lung cancer

Associated Data

Supplementary Materials
Data Availability Statement

Abstract

Immunotherapy utilizing checkpoint inhibitors has shown remarkable success in the treatment of cancers. In addition to immune checkpoint inhibitors, immune co-stimulation has the potential to enhance immune activation and destabilize the immunosuppressive tumor microenvironment. CD137, also known as 4-1BB, is one of the potent immune costimulatory receptors that could be targeted for effective immune co-stimulation. The interaction of the 4-1BB receptor with its natural ligand (4-1BBL) generates a strong costimulatory signal for T cell proliferation and survival. 4-1BBL lacks costimulatory activity in soluble form. To obtain co-stimulatory activity in soluble form, a recombinant 4-1BBL protein was generated by fusing the extracellular domains of murine 4-1BBL to a modified version of streptavidin (SA-4-1BBL). Treatment with SA-4-1BBL inhibited the development of lung tumors in A/J mice induced by weekly injections of the tobacco carcinogen NNK for eight weeks. The inhibition was dependent on the presence of T cells and NK cells; depletion of these cells diminished the SA-4-1BBL antitumor protective effect. The number of lung tumor nodules was significantly reduced by the administration of SA-4-1BBL to mice during ongoing exposure to NNK. The data presented in this paper suggest that utilizing an immune checkpoint stimulator as a single agent generate a protective immune response against lung cancer in the presence of a carcinogen. More broadly, this study suggests that immune checkpoint stimulation can be extended to a number of other cancer types, including breast and prostate cancers, for which improved diagnostics can detect disease at the preneoplastic stage.

Supplementary Information

The online version contains supplementary material available at 10.1007/s00262-023-03507-2.

Keywords: 4-1BB, SA-41BBL, Cancer immunotherapy, Lung cancer, NNK, Immunomodulation

Introduction

Cancer is a major global public health problem and the second leading cause of death worldwide [1]. Around 10 million people die annually from cancer, and the number of cases is expected to roughly triple over the next two decades [2]. It has been demonstrated that cancer cells can be identified and eliminated prior to tumor formation by immune surveillance mechanisms [3] which includes communication between adaptive and innate immune cells. To escape the immune surveillance system and promote tumor growth, tumor cells evade immune system attacks through various mechanisms such as restricting antigen recognition, creating an immunosuppressive environment to suppress the immune system, and causing T cell exhaustion [4].

In 1891, William B. Coley showed the efficacy of immune stimulation as immunotherapy by injecting bacteria into a cancer patient in order to boost the immune system for tumor elimination.

Since then, the understanding and application of immunotherapy as a cancer treatment has advanced significantly. Numerous immunotherapeutic drugs have been investigated in preclinical models, and those that have demonstrated efficacy, such as immune checkpoint inhibitors, have been granted regulatory approval [3]. In recent years, the clinical usage of anti-PD1 (Nivolumab) and anti-CTLA-4 (Ipilimumab) checkpoint inhibitors has emerged with significant success in the treatment of malignancies [5]. In addition, immune co-stimulation to immune checkpoint inhibitors holds great potential to boost immune activation and destabilize the immunosuppressive tumor microenvironment.

CD137, also known as 4-1BB, is one of the potent immune costimulatory receptors that could be targeted for effective immunomodulation. 4-1BB, a member of the tumor necrosis factor (TNF) receptor family, is expressed on the surface of activated T, B, and NK lymphoid cells. Signaling via 4-1BB enhances the survival, proliferation, and differentiation of T cells into effector cells and the generation of long-term memory [68]. Activation of the 4-1BB receptor has been shown in preclinical studies to enhance the antitumor activity of T cells and increase the efficacy of cancer immunotherapy. With promising findings, this strategy has also been evaluated in clinical trials to assess the efficacy of agonistic 4-1BB antibodies alone or in combination with other immunotherapy and chemotherapy regimens. However, clinical trials with anti-4-1BB agonistic antibodies failed due to unacceptable systemic toxicity [9, 10].

Contrary to agonistic antibodies, 4-1BBL, the natural ligand for the 4-1BB receptor, does not induce toxicity upon binding to the 4-1BB receptor. 4-1BBL is a cell surface membrane protein that is expressed on dendritic cells, macrophages, B cells, and it is active only in its membrane-bound form. The trimeric (human) or dimeric (mouse) soluble form of 4-1BBL requires crosslinking to cluster the 4-1BB receptors for efficient signaling to immune cells [1113].

Therefore, to construct a functional soluble natural ligand for the 4-1BB receptor, we fused the extracellular domain of murine 4-1BBL to a modified form of the streptavidin core (SA) to make a chimeric molecule (SA-4-1BBL). Due to the structural characteristics of SA, chimeric SA-41BBL formed tetramers and oligomers [14, 15]. Soluble SA-4-1BBL exhibited significant immunological activity in T effector cell activation, acquisition of effector functions, and long-term memory formation [16]. We previously reported that SA-4-1BBL prevented the conversion of T-conventional cells into T-regulatory cells, which was triggered by IFNγ production in T-conventional cells [17], and overcame Treg suppression through inducing the production of IL-2 by T-effector cells [14]. Furthermore, unlike 4-1BB antibodies, we did not observe systemic toxicity with repeated administration of SA-4-1BBL in mice [13]. Importantly, when used as an adjuvant in tumor-associated antigen-based subunit vaccines, SA-4-1BBL demonstrated therapeutic activity in a variety of preclinical tumor models [15, 16, 1821]. We recently reported that mice treated with soluble SA-4-1BBL protein were protected against a challenge with transplantable tumors such as Lewis lung carcinoma (LLC), TC-1, and 3LL-huMUC1 [22]. Regardless of tumor type, this effect was long-lasting and required CD4 T cells, NK cells, and IFN-γ. In addition, treatment with SA-4-1BBL after surgical removal of tumors prevented recurrences [22].

Here, we demonstrated that the immunotherapeutic potential of SA-4-1BBL is not restricted to transplantable tumor models, but also extends to carcinogen-induced lung cancer in a rodent model with high susceptibility to carcinogen-induced tumors.

The data presented in this paper showed that utilizing an immune checkpoint stimulator as a single agent generated a protective immune response against lung cancer in the presence of carcinogen. A deeper understanding of the molecular basis of immune checkpoint stimulation may also offer opportunities to combine this therapy with potentially additive or synergistic biologics for enhanced immunological efficacy. Notably, this immunotherapeutic platform can be extended to a number of other cancer types, including breast and prostate cancers, for which improved diagnostics can detect disease at the preneoplastic stage.

Materials and methods

Animal studies

The animal study protocol was approved by the Animal Care and Use Committee of the University of Missouri-Columbia (IACUC–9893). Female A/J mice (18–22 g) were obtained from The Jackson Laboratory (Stock #000646; Bar Harbor, ME, JAX) and housed in plastic cages with free access to water and a basal diet under controlled conditions of 22 °C and 45% humidity in a 12-h light–dark cycle. The mice were randomly divided into groups, as detailed in the experimental design.

NNK administration

NNK (4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone) was purchased from Santa Cruz Biotechnology (Cat#: sc-209854) and dissolved in DMSO (dimethyl sulfide; Sigma, Cat#: D2650). Injection doses of NNK (25 mg/kg) were prepared in sterile saline and administered intraperitoneally each week for eight consecutive weeks.

SA-4-1BBL protein administration

As previously described [14], SA-4–1BBL was manufactured in our laboratory following established protocols. At the sixth and eighth week, 48 h after i.p. injection of NNK, mice were subcutaneously injected with 100 µg of SA-4-1BBL. Mice in the NNK control group were injected with vehicle (sterile saline).

Lung tissue sectioning, hematoxylin and eosin staining, and tumor nodule evaluation

Animals were sacrificed, and their lungs were harvested at the end of the study (18 weeks). Using a dissecting microscope, two trained investigators evaluated the whole lung for lung macroscopic nodules in a blinded manner. Macroscopic tumor nodules with a bright pink color were located on the lung surfaces, making them easily distinguishable from the surrounding lung tissues. After assessing the lungs for macroscopic tumor formation, a portion of each lung lobes was drained, weighed, and fixed with 10% neutral-buffered formalin for histopathology. To evaluate microscopic tumor formation, histopathological analyses were performed. The lung tissues were fixed in formalin for 48 h, paraffin-embedded and cut as 8 µm/section using a Leica RM2235 microtome. Sequential tissue sections were taken 40 µm apart for the entire block. A minimum of 60 sections per mouse were stained with Hematoxylin and Eosin (H&E) to evaluate microscopic tumor nodule and calculate the number of tumor nodules per animal. The presence of proliferative lung lesions was evaluated according to the Mouse Models of Human Cancers Consortium (MMHCC) [23]. Lung tumors were not sub-classified into adenoma and adenocarcinoma due to limitation of immunohistopathology analysis to distinguish malignancy after 16 weeks post-NNK injection [24]. H&E-stained lung tissues were analyzed using an Olympus CKX53 inverted microscope. Digital computer images were recorded with an EVOS M7000 imaging system. 10X optical magnification was used to acquire EVOS digital microscopy images, and 10X, 20X, and 40X digital magnification was used to generate total magnifications of 100X, 200X, and 400X. Repeated nodules accepted as one nodule through examining the sequential sections in microscopic tumor examination. Evaluation of the microscopic tumor nodules was conducted in a blinded manner. The total microscopic tumor numbers were recorded for each animal, and statistical analysis was performed with a one-way ANOVA test in GraphPad Prism v.9.

Microscopic tumor nodules area analysis

The microscopic tumor area was measured from H&E-stained lung sections. Five lung samples per treatment group were analyzed. The three largest mouse tumor nodules were identified and an EVOS M7000 microscope (Invitrogen) was used to mark the margins of the tumor nodules. The, dimensions of the tumor nodule and the total lung section area were measured using EVOS Analysis software. The total area of tumor nodules per lung section was calculated and presented as a percentage of total lung area. For statistical comparison, a minimum of 15 lung nodules per group were included and statistical difference calculated using a one-way ANOVA test. (GraphPad Prism v.9).

Immunofluorescence staining

For immunofluorescence staining, proliferating cell nuclear antigen (PCNA) antibody was used to show the proliferative capacities of tumors seen in H&E-stained slides. PCNA is a nuclear non-histone protein that takes a role during DNA synthesis. Therefore, it is one of the most frequently used proliferation markers for analyzing different neoplasms and pathological grading in the clinic and preclinical studies [25, 26]. Processed paraffin sections were deparaffinized in xylene (Fisher, Cat#: 22-143,975), rehydrated through graded ethanol-H2O solutions, and washed in distilled water. Antigen retrieval was done by treating sections in 10-mM sodium citrate (Fisher Chemical, Cat#: BP3271) buffer (pH 6.0) for 15 min in a boiling water bath and washed with phosphate-buffered saline (PBS). Nonspecific binding sites were blocked by incubation with 5% bovine serum albumin (BSA, Sigma, Cat#: A70300), 0.3% Triton-X (Sigma, Cat#: T8787) dissolved in PBS, 2.5% normal goat serum (ImmunoReagents, Inc., Cat#: SP-004-VX2), and Fc block (BD Pharmingen, Cat#: 553,142, Clone: 2.4 G2). Sections were incubated overnight at 4 °C with PCNA primary antibody (Novus Bio, Cat#: NB100-456, 1:100 dil.). Then, tissue sections were washed with PBS + 0.05% Tween-20 (Sigma-Aldrich, Cat#: P5927) and incubated with secondary antibody Alexa-Fluor 647 (Life Technologies, Cat#: A21244, 1:150 dilution) for 1 h at room temperature before counterstaining with 5 µM SYTOX blue nucleic acid stain (Fisher Scientific, Cat#S11348). After washing with distilled water, slides were covered with Immu-Mount fluorescent mounting media (Epredia, Cat#: 9,990,402). Stained slides were imaged using a Leica TCS SP5 confocal microscope.

In vivo immune cells depletion

Antibodies were administered intraperitoneally (i.p.) to deplete CD4 (clone GK1.5, 500 µg/injection), CD8 (clone 53.6.72, 500 µg/injection), and NK1.1 (clone PK136, 500 µg/injection) immune cell populations at weeks six and eight of NNK treatment. The control group was given an i.p. vehicle (sterile saline). Flow cytometric analysis confirmed depletion in peripheral blood at 72-h and 12-week post-antibody injection. Anti-CD3-V500 (BD Bioscience, Cat#: 560,771), anti-CD4 Alexa-Fluor 700 (BD Pharmingen, Cat#: 557,956, clone RM4-5), anti-CD8-APC-Cy7 (BD Pharmingen, Cat#: 557,654), anti-NK1.1-APC (BD Bioscience, Cat#: 550,627) and 7AAD (BD Pharmingen, Cat#: 51-68981E) fluorescent-conjugated antibodies were used to assess depletion of CD4+, CD8+ and NK1.1+ cells across the experimental groups.

Flow cytometry and immunophenotyping

Lung tissues were mechanically dissociated and digested with 0.5 mg/ml Type IV Collagenase (Worthington Biochemical, Cat#: 4186) by constant shaking (New Brunswick Scientific, I-24 incubator Shaker Series) for 40 min in 37[degree celsius]. Digested lung tissues were filtered using 40-µm nylon cell strainer (pluriStrainer, Cat#: 43-50,050-01) to elute single cells. Lung-draining lymph nodes were mechanically dissociated, and sample were passed through an 18 G 1 ½ needle (BD PrecisionGlide Needle, Cat#: 305,195) attached to a 10-cc syringe (BD, Cat#: 303,134), to make a single cell suspension. The single cell suspension was filtered with a 100-µm nylon mesh (Elko, Cat#: 03–100/32). Lung and lung-draining lymph node single-cell isolates were washed with PBS twice and counted using trypan blue dye for viable cell counting. 1 million lung-draining lymph node cells and 2–3 million lung infiltrate cells per mouse were incubated with Mouse BD Fc Block (anti-mouse CD16/CD32; BD Pharmingen, Cat#: 553,142, Clone: 2.4 G2) at 4 °C in dark for 15 min. Lung infiltrating lymphocytes and lung draining lymph node cells were subsequently stained for cell surface markers for 30 min (antibody panel outlined in Supplementary Table S1). Cell subpopulations were identified using a Cytek Aurora flow cytometer by gating on the live CD45+ cell population. FCS Express v.7 was used to compute cell percentages and absolute cell numbers. Absolute cell numbers and cell percentages were analyzed using a one-way ANOVA. (GraphPad v.9).

Statistics

Statistics were performed with GraphPad Prism v.9 software (La Jolla, CA). Student's t tests and one-way ANOVA were used to determine significant differences between groups. In addition, two-way ANOVA was used to evaluate the effects of two variables, treatment (NNK v.s. NNK + SA-4-1BBL) and time. A two-way ANOVA was also used to assess the body weight changes between different treatment groups. All p-values  0.05 were considered statistically significant.

Results

Treatment with NNK-induced lung tumorigenesis

To evaluate the immunotherapeutic efficacy of SA-4-1BBL in genetically predisposed model [27], we used A/J mice because of their susceptibility to nicotine derivative-induced lung cancer, making them one of the best preclinical animal models for smoking-related lung cancer [2831]. A/J mice are predisposed to spontaneous lung tumor development due to a mutation in the PAS1 gene, which is linked to the KRAS protooncogene [32]. The KRAS protooncogene regulates cell proliferation and division, and mutations in this gene are associated with the development of numerous cancers, including lung cancer. We used NNK (4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone), which is the most carcinogenic nitrosamine nicotine derivative in humans and animals, to accelerate tumor development [2830]. Lung cancer development was accelerated in A/J mice by injecting NNK at a weekly dose of 25 mg/kg for eight weeks [27]. In the sixth- and eighth-week post-NNK injections, SA-4-1BBL protein was administered subcutaneously (Fig. 1A). Animals were assessed each week for 18 weeks to determine whether there were any differences in the animals' wellbeing after receiving NNK and SA-4-1BBL treatments. Body weight evaluation showed no significant differences across groups compared to untreated age-matched A/J mice (Fig. 1B). At the end of the study, necroscopy was performed to detect any NNK and/or SA-4-1BBL treatment-related adverse pathological changes in internal organs such as the kidney, liver, bladder, and stomach. No measurable differences were observed between the treatment and control groups (data not shown), which was consistent with the previously reported safety profile of SA-4-1BBL treatment [13, 22].

An external file that holds a picture, illustration, etc.
Object name is 262_2023_3507_Fig1_HTML.jpg

SA-4-1BBL treatment protected mice from developing lung tumors induced by NNK. A Experimental design, and treatment timelineFemale A/J mice were randomly divided into three groups: naïve (untreated), NNK control, and NNK + SA-4-1BBL-treated group. NNK control (NNK + vehicle) and NNK + SA-4-1BBL groups received 25 mg/kg of NNK (4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone) via i.p. injection weekly for the first eight weeks of the experiment. SA-4-1BBL treatment group (n = 17) also received 100-µg SA-4-1BBL s.c. at weeks 6 and 8, two days post-NNK administration. The animals body weight changes were assessed during weekly follow-up. Body weight changes were not significantly different across the groups compared to untreated age-matched A/J mice. C. Representative images of macroscopic tumor nodules in the lungs. In the 18th week, mice were euthanized, and lungs were perfused to evaluate macroscopic tumor nodules. Black dashed circles show macroscopic tumor nodules on the lungs. D. Macroscopic tumor numbers in NNK control and NNK + SA-4-1BBL treatment groups. Lungs were evaluated under the dissecting microscope, and macroscopic tumor nodules were counted and compared between the NNK control group and NNK + SA-4-1BBL treatment group. SA-4-1BBL-treated group showed significantly fewer macroscopic tumor nodules than the control group (p < 0.01). E. Microscopic tumor nodule numbers in NNK control and NNK + SA-4-1BBL treatment groups. Lungs were sectioned and stained with H&E for immunohistochemistry analysis. NNK + SA-4-1BBL-treated group had significantly fewer microscopic tumor nodules than the NNK control group (p < 0.0001). F. Representative images of sections containing tumor nodules stained with H&E and PCNA. Images in the first row (a and b) illustrate hyperplastic cell structures with increased nucleus-to-cytoplasm ratio in H&E-stained lung tissue (red dashed circle). Representative images were captured using EVOS M7000 Imaging System (ThermoFisher Scientific) with employing a 10X optical magnification. H&E-stained (a) and (b) images presented at 100X and 200X total magnification, with 500-µm and 100-µm scale bars, respectively. Lung sections were stained with proliferation marker PCNA antibody. PCNA staining (red) and SYTOX blue nuclear staining (blue) are depicted in the second row (c and d) captured with 20X objective in Leica confocal microscopy. Scale Bars: 100 µm and 25 µm for NNK + vehicle group (with zoom factor 1 and 1.35) and 100 µm and 75 µm for NNK + SA-4-1BBL (with zoom factor 1 and 1.56). The tumor area (yellow dashed circle) had a higher PCNA staining profile than the alveolar lung tissues surrounding the tumor area. The data presented in this figure were compiled from three to four independent experiments involving four to six mice per experiment and are shown as mean ± standard deviation (SD). Body weight changes were analyzed using two-way ANOVA B, macroscopic D and microscopic E tumor nodule numbers were analyzed by unpaired students’ t test

SA-4-1BBL as monotherapy significantly decreased the number of tumor nodules in NNK-induced lung cancer

In our previous study, pretreatment with SA-4-1BBL, as an agonist of the 4-1BB costimulatory pathway, protected mice from subcutaneous tumor challenge without antigen priming. In this study, we investigated whether treatment with SA-4-1BBL protein inhibits tumor growth in a carcinogen-induced mouse model of lung cancer. A/J mice were injected with 100 µg of SA-4-1BBL at weeks 6 and 8 of NNK injections (Fig. 1A). At the end of the study (18 weeks), mice were euthanized, and two trained investigators counted macroscopic tumor nodules on the lung surfaces using dissecting microscope. Mice given SA-4-1BBL protein had significantly fewer macroscopic tumor nodules than the control group (NNK + vehicle) (Fig. 1C and D). The tumor prevention capacity of SA-4-1BBL was consistently seen in repeated experiments, further demonstrating the power of immune checkpoint stimulation. Visible tumor nodules were primarily observed in the periphery of the lungs (Fig. 1C, black circles), which is similar to smoking-related lung cancer in humans.

The macroscopic tumor nodules were further confirmed as cancerous lung nodules using histopathological analyses. Sectioned lung tissues were stained with H&E and tumor nodules were counted microscopically by two trained investigators in a blinded manner. PCNA immunofluorescence labeling was used to confirm the malignant phenotype with proliferative capacities of microscopic tumor foci defined by H&E staining (Fig. 1F). Mice treated with SA-4-1BBL had significantly fewer microscopic tumor foci than the control group (Fig. 1E). Moreover, one animal in the SA-4-1BBL treatment group showed no microscopic lung tumors. A strong significant reduction in the number of microscopic tumors may be attributable to the increased efficacy of SA-4-1BBL treatment on tumor nodules that have already formed prior to obtaining their first dose of SA-4-1BBL. Tumor nodule size was also measured by examining microscopic tumor areas to further analyze the proliferative capacity of the tumor cells associated with NNK exposure in A/J mice (Fig. 3A and C). Notably, both the tumor dimensions and the percentage of tumor area in lung sections in the SA-4-1BBL treatment group were significantly smaller than those in the control group (Fig. 3C and D).

An external file that holds a picture, illustration, etc.
Object name is 262_2023_3507_Fig3_HTML.jpg

T and NK cell depletion impaired the immunotherapeutic efficacy of SA-4-1BBL. As previously described, female A/J mice received weekly doses of NNK for eight weeks, followed by two doses of SA-4-1BBL treatment and depletion antibodies. In the experimental endpoint, lung tissues were sectioned, and H&E-stained slides were used to assess microscopic tumor size changes within different treatment groups. Microscopic tumor nodule margins were defined, and digital microscopy images were captured at 10X magnification. A. Representative microphotograph of H&E-stained lung tissue samples from naïve A/J mice, control group (NNK + vehicle), and NNK + SA-4-1BBL-treated group. The area marked by the black line shows microscopic tumor nodules in lung from the NNK control group. B. H&E-stained lung tissue samples from groups treated with SA-4-1BBL and then depleted of CD4+ T cells, CD8+ T cells, or NK cells. Microscopic tumor nodules marked with black lines. The EVOS M7000 Imaging System (ThermoFisher Scientific) was used to acquire representative microscopy images in A and B, at 100X, 200X, and 400X total magnification, with 500-µm scale bars, employing a 10X optical magnification. Variations in the dimensions of the embedded tissue resulted in non-uniform images when sections were magnified uniformly to compensate for ratio differences between sections. C. A representative image of tumor nodule dimension and total lung section dimension measurement in the H&E-stained section (left), and a comparison of microscopic tumor nodule areas between the groups (right). SA-4-1BBL-treated group microscopic tumor nodule dimensions were significantly smaller than the NNK control group and immune cell-depleted groups (n = 15 tumor nodules/group, p < 0.05 and p < 0.01). D. The percent of the tumoral areas over the non-tumoral lung areas (n = 15 tumor nodules/group, p < 0.05). Tumor area dimension and the percent tumor areas to the lung section area in the SA-4-1BBL treatment group were significantly smaller than those in the control group. A p-value  0.05 was considered significant. Microscopic tumor dimensions C and tumor area percentages D were compared using a one-way ANOVA. Data represented as mean ± standard deviation (SD). Scale bars, 500 µm

SA-4-1BBL-mediated tumor prevention depended on CD4, CD8, and NK cells

In our reported study, the tumor preventive efficacy of SA-4-1BBL was shown to be dependent on cross-communication between CD4+CD44high “memory-like” T cells and NK cells, with a requirement for IFN-γ [22]. A series of depletion experiments were conducted to determine the function of immune cell populations in mediating the immunotherapeutic efficacy of SA-4-1BBL in a NNK carcinogen-induced tumor model. Depleting antibodies against CD4+, CD8+ and NK cells were injected i.p. following SA-4-1BBL treatment during the sixth and eighth weeks of NNK administration (Fig. 2A). Immune cell depletion showed no adverse effects on the health of mice, including no differences in body weight between groups (Fig. 2B).

An external file that holds a picture, illustration, etc.
Object name is 262_2023_3507_Fig2_HTML.jpg

In vivo depletion of CD4+, CD8+, and NK cells abolished SA-4-1BBL antitumor activity. A. Experimental design; NNK administration, SA-4-1BBL treatment, and in vivo depletion timeline. Female A/J mice were randomly divided into groups, and all groups received weekly 25 mg/kg NNK injections for eight weeks. In the sixth and eighth weeks of NNK injections, selected groups received SA-4-1BBL injection subcutaneously. Twenty-four hours after SA-4-1BBL treatment, antibodies against different immune cell populations were injected to deplete CD8+ cells (n = 6), NK cells (n = 6), or CD4+ cells (n = 9). B. The animals body weight changes were assessed during weekly follow-up. Body weights were not significantly different in the SA-4-1BBL treatment and SA-4-1BBL + depletion groups compared to the control group. C. Representative images of macroscopic tumor nodules on the lung surface. Lungs were harvested, and macroscopic tumor numbers were assessed under a dissecting microscope at the experimental endpoint. Black dashed circles show macroscopic tumor nodules in the lungs of the mice from the NNK control group, CD4+ T cell, CD8+ T cell, and NK cell-depleted groups. D. Macroscopic tumor numbers between different treatment groups. Counted macroscopic tumor numbers were compared between the groups. The NNK + SA-4-1BBL-treated group (n = 17) has significantly fewer macroscopic tumor nodules than the NNK control group (n = 16) and CD4+ T cells depleted group (n = 9, p < 0.05). E. Lung tissue sections were stained with H&E, and microscopic tumor nodules were enumerated from each group. Microscopic tumor formation compared between NNK control (n = 15), NNK + SA-4-1BBL treatment group (n = 15), SA-4-1BBL + CD4 T cell-depleted (n = 8), SA-4-1BBL + CD8+ T cell, and SA-4-1BBL + NK cells depleted groups (n = 6). SA-4-1BBL-treated group had significantly fewer microscopic tumor nodules in the lungs (p < 0.001) as compared to NNK control and depletion groups. The data presented in this figure were compiled from three to four independent experiments. A p-value  0.05 was considered significant. Data are represented as mean ± standard deviation (SD). Body weight changes were analyzed using a two-way ANOVA B, and a one-way ANOVA was used for analysis of macroscopic D and microscopic E tumor numbers

CD4+ T cell, CD8+ T cell and NK cell depletion abrogated the protective antitumor effect of SA-4-1BBL treatment. In mice depleted of CD4+ T cells (Supplementary Fig. S1A), there was a significant increase in both macroscopic and microscopic tumor nodules compared to mice that did not receive CD4+ T cell depleting antibody (Fig. 2C, D, E). In contrast, depletion of CD8+ T (Supplementary Fig. S1A) cells and NK cells resulted in a significant increase in microscopic tumor nodules, but no significant change was seen in macroscopic tumor nodule numbers (Fig. 2C, D, E). These results indicate that CD4+ T cell help is required for the generation of antitumor primary immune responses, which is consistent with our previous study [22].

The tumor burden was restricted to the lung, as detailed evaluation of organs using a dissecting microscope showed none of the mice in the study groups had tumor nodules in other organs. Notably, one mouse in CD4+ T cell-depleted group had a tumoral lesion on its chin. This lesion was also removed on the day of termination, and histopathologic analysis was performed. The analysis revealed that the tumor was metastatic, and it had the same histological phenotype as the tumors seen in the lung (Supplementary Fig. S2A-C). This metastatic nodule had 14-fold more PD1+CD4+ and PD1+CD8+ cells (PD-1, programmed cell death protein 1) than the lung tumors (Supplementary Fig. S2D and S2E). In contrast to previously published transplantable tumor models, depletion of CD8+ T cells had a significant effect on the antitumor activity of SA-4-1BBL in a NNK carcinogen-induced model [22]. These results suggest that CD8+ T cells play an essential role in the antitumor activity of SA-4-1BBL in the carcinogen-induced lung tumor (Fig. 2C and andE).E). As compared to SA-4-1BBL-treated controls, tumor areas were significantly increased in the lungs of SA-4-1BBL-treated mice depleted of either T or NK cells and were comparable to the NNK control (Fig. 3B, B,C,C, ,D).D). These data further confirmed that the immunotherapeutic effectiveness of SA-4-1BBL treatment in inhibiting tumor growth was dependent on the presence of T and NK cells.

Overall, these findings provided important insights into the mechanisms underlying the immunotherapeutic effects of SA-4-1BBL treatment and highlighted the importance of T cells and NK cells in the immune response against cancer.

Treatment with SA-4-1BBL increased the number of naive and central memory CD4+ T cells and decreased the number of CD8+PD-1+ cells

We have previously shown that pretreatment with SA-4-1BBL elicited a long-lasting immune memory response, which was predominantly mediated by NK cells and CD4+ T cells expressing IFN-γ [22]. Here, we utilized deep immune phenotyping to assess the immune cell compositions in the lung and draining lymph nodes. SA-4-1BBL treatment resulted in a significant increase in the absolute cell number of CD4+ central memory T cells (CD4+CD44hiCD62Lhi) in lung and lung draining lymph nodes compared to the NNK control group (p < 0.05) (Fig. 4A, B). The increased number of central memory T cells in SA-4-1BBL-treated animals suggested that these cells were traveling between the lung and the lung-draining lymph nodes, where they had encountered antigen, been activated, and then migrated to lymphoid tissues where they survived for extended periods. The number of naive CD4+ T lymphocytes (CD4+CD44loCD62Lhi) in the lung-draining lymph node of SA-4-1BBL-treated mice was also significantly higher, which may serve as a constant source of central memory cells and provide long-term protection against tumor growth (Fig. 4C). In addition, there was a significant decrease in the number of PD-1 positive CD8+ T cells in the draining lymph node of mice treated with SA-4-1BBL (Fig. 4D). PD-1 is an inhibitory receptor expressed on the T cell surface. The ligand of PD-1, named as Programmed Death-Ligand 1 (PD-L1), is found on the surface of many different cells, including cancer cells. When PD-L1 binds to PD-1, an inhibitory signal is transmitted that inhibits T-cell activation, thereby dampening the immune response. In preclinical models, it has been demonstrated that NNK-induced lung tumors express PD-L1 [28, 29]. The reduction in PD-1-positive CD8+ T cells could be an additional mechanism by which SA-4-1BBL exerts its antitumor growth effect. However, further research is needed to confirm this hypothesis and fully understand the role of SA-4-1BBL in modulating the immune responses against cancer in another induced or spontaneous tumor model.

An external file that holds a picture, illustration, etc.
Object name is 262_2023_3507_Fig4_HTML.jpg

SA-4-1BBL treatment increased the number of naive and central memory CD4+ T cells but decreased CD8+PD-1+ cells. Deep immune phenotyping was performed on lung tissue and lung draining lymph node at the experimental endpoint. A. SA-4-1BBL treated group (n = 10) had significantly higher central memory CD4+ T cell numbers (CD4+CD44hiCD62Lhi) in the lung, and B. lung-draining lymph nodes compared to NNK control group (n = 10, p < 0.05), and depletion groups (n = 6–7, p < 0.001). C. SA-4-1BBL treatment (n = 10) increased naïve CD4+ T cell (CD4+CD44loCD62Lhi) in the lung draining lymph nodes, compared to control groups (n = 10, p < 0.001), and immune cell-depleted groups (n = 6–7, p < 0.1). D. SA-4-1BBL treatment (n = 10) significantly decreased the number of CD8+PD-1+ cells in lung-draining lymph nodes, compared to control group mice (n = 10, p < 0.05). The data presented in this figure were compiled from three to four independent experiments. A p-value  0.05 was considered significant. Absolute cell numbers were analyzed using a one-way ANOVA A, B, C and D. Data represented as mean ± standard deviation (SD)

Discussion

Immune checkpoint inhibitors such as anti-CTLA-4 and anti-PD-1/PD-L1 have been shown to restore T cell-mediated antitumor immunity and induce durable remissions in some patients with cancer. The FDA (Food and Drug Administration) has approved these treatments for the treatment of several types of cancer, including melanoma, lung cancer, bladder cancer, kidney cancer, head and neck cancer, and Hodgkin lymphoma [5, 3034]. However, these treatments are not effective for all types of cancer, and not all patients respond equally well [3538]. Therefore, there is a need for robust immune stimulation to obtain higher response rates and long-term remissions. Combining checkpoint inhibitors with robust immune co-stimulation thus becomes an attractive strategy.

One approach to achieving robust immune stimulation is combining two or more immunotherapeutic agents that can target different aspects of the immune system and work in synergy to potentially increase the antitumor immune response [39, 40]. Combining immune checkpoint inhibitors with other immunotherapeutic agents such as cancer vaccines, adoptive cell therapy, or immune stimulators has shown promising results in preclinical and clinical studies. In some cases, combination therapy has demonstrated improved response rates and longer survival than single-agent therapy [41, 42].

The 4-1BB/4-1BBL pathway has been shown to have a critical role in the regulation of immune responses[43]. Upon antigenic stimulation, such as exposure to a pathogen or tumor cells, the expression of 4-1BB receptor is upregulated on immune cells, including T cells and natural killer cells. The membrane-bound form of 4-1BBL binds to the 4-1BB receptor to provide a potent costimulatory signal that enhances immune cell activation, proliferation, and survival. Activation of this pathway has been shown to stimulate immune responses, particularly in the context of cancer immunotherapy [43, 44].

The soluble form of 4-1BBL has limited immunostimulatory activity because efficient signal transduction requires receptor crosslinking for multimerization. To generate a stable soluble form of 4-1BBL with potent co-stimulatory activity, we generated a chimeric molecule comprised of the extracellular portion of murine 4-1BBL and streptavidin (SA-4-1BBL). SA-4-1BBL, a novel form of 4-1BBL, forms tetramers and oligomers [14, 15] with high immunological potential to activate T effector cells. The soluble SA-4-1BBL has been shown to have antitumor effects in preclinical studies [1315, 18, 20, 22, 45] with no detectable toxicities as compared to 4-1BB agonistic antibodies.

The aim of this study was to determine whether SA-4-1BBL could be used as an immune checkpoint stimulator in mice predisposed to spontaneous cancer development. For this purpose, SA-4-1BBL was administrated to A/J mice during ongoing exposure to tobacco carcinogens NNK, to determine whether treatment with SA-4-1BBL could halt or reverse the incidence of lung tumors compare to the group that did not receive SA-4-1BBL treatment. We adopted a previously described NNK-induced lung cancer model by administering intraperitoneal injections of 25 mg/kg NNK carcinogen weekly for eight weeks. We demonstrated that eight weekly injections of NNK were sufficient to induce smoking-related lung cancer by 18 weeks with 100 percent tumor incidence, compared to the intragastric gavage method, which requires 30 to 40 weeks to develop lung tumor nodules. The dose of SA-4-1BBL protein used in this study was based on our previous reported study, in which 100 µg was the optimal dose to protect mice from subsequent subcutaneous tumor challenges, and lower protein doses provided compromised protection compared to 100 µg [22]. Although we tested higher protein doses in A/J mice, the most efficacious regimen consisted of two 100-µg doses administered two weeks apart.

We herein report that the therapeutic doses of SA-4-1BBL administered to A/J mice did not result in detectable macroscopic toxicity, which was consistent with numerous preclinical cancer models reporting the same therapeutic doses without toxicity [13, 15, 18, 20, 22, 45].

Treatment of A/J mice, which are genetically predisposed to spontaneous cancer development, with the SA-4-1BBL protein during active carcinogen exposure prevented the development of lung tumor nodules. This prevention was significant compared to NNK carcinogen-exposed mice without SA-4-1BBL treatment which developed a higher number of macroscopic and microscopic tumor nodules (Fig. 1C, C,D,D, ,E,E, ,F).F). Furthermore, the depletion of CD4+, CD8+ T cells, and NK cells significantly impaired the antitumor activity of SA-4-1BBL, indicating that the therapeutic effect of SA-4-1BBL was mediated by these immune cells (Fig. 2). Mice depleted of CD4+ T cells had an increase in the number of both macroscopic and microscopic tumor nodules. In contrast, the depletion of CD8+ T and NK cells increased the number of microscopic tumor nodules significantly but had no effect on the number of macroscopic tumor nodules (Figs. 2 and and3).3). These findings suggested the antitumor activity of SA-4-1BBL immunotherapy is primarily mediated by CD4+ T cells, with active participation from CD8+ T and NK cells. These results also indicated a cross-communication between innate and adaptive immunity. It is well-known that CD4+ T cells play a crucial role in initiating an active immune response as they help activate other immune cells and boost antibody formation. CD8+ T and NK cells, on the other hand, are directly responsible for destroying infected or malignant cells. In the absence of these cells, the immune system's ability to target and eliminate malignant cells are compromised. Consistent with a previously published study [22], SA-4-1BBL treatment significantly increased CD4+ naive and central memory cell numbers in the lung and lung draining lymph node (Fig. 4A, B, C). In response to antigen exposure, naive CD4+ T cells become activated and differentiate into effector T cells, which can migrate to the site of an infection or tumor to eradicate the pathogen or tumor cells. Some of these effector T cells differentiate into memory T cells, including central memory T cells, which are able to persist in the target tissues and provide long-term protection against subsequent infections with the same pathogen or tumor. These cells might be the cells exposed to tumor antigens and further stimulated by SA-4-1BBL therapy, although their exact nature has not yet been fully determined. Treatment with SA-4-1BBL decreased the number of PD-1-expressing CD8+ T cells compared to NNK-exposed control mice that were not treated with SA-4-1BBL (Fig. 4D). PD-1 is expressed on the surface of activated T cells, including CD8+ T cells, and regulates the immune response. Chronic antigen exposure, such as during prolonged infections or in the presence of cancer cells, can induce the expression of PD-1, leading to T cell exhaustion, which limits T cells’ capacity to eliminate target cells. Therefore, a significant reduction in the number of CD8+ T cells expressing PD-1 suggests that SA-4-1BBL may enhance the immune response against NNK-induced cancers by reducing T cell exhaustion. It is important to note that without additional data, a complete interpretation of the results cannot be made.

In conclusion, our findings demonstrated the immunomodulatory effect of SA-4-1BBL during continuous carcinogen exposure in mice predisposed to cancer. These findings provide additional evidence that immunotherapy utilizing potent immunostimulatory molecules, such as SA-4-1BBL, has the potential to treat and prevent cancer by enhancing the immune system's ability to identify and eliminate cancer cells. Thus, co-stimulation-based immunotherapy can provide precise, targeted treatment that avoids many of the adverse side effects associated with chemotherapy and radiation in various cancer treatments.

Supplementary Information

Below is the link to the electronic supplementary material.

Acknowledgements

The authors thank The NextGen Precision Health Building, at the University of Missouri-Columbia for providing advanced core facilities. The authors also thank Dr. Christa Jackson for her critical reading of the manuscript

Abbreviations

H&EHematoxylin and eosin
IHCImmunohistochemical staining
NNK4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone
PCNAProliferating cell nuclear antigen
DMSODimethyl sulfide
SAStreptavidin
s.c.Subcutaneous injection
i.p.Intraperitoneal injection
PD-1Programmed cell death protein-1
PD-L1Programmed death-ligand 1
IFNγInterferon γ

Author contributions

Conception and design were done by ESY and HS. AEG, RR, MT and VU performed the experiments. AEG, RR and MT analyzed data and prepared Figs. 1, ,2,2, ,33 and and4.4. ESY, HS and AEG wrote the manuscript. All authors reviewed the manuscript.

Funding

This study was supported by the Department of Defense (DoD) grant number LC190524.

Data availability

SA-4-1BBL protein is available through a material transfer agreement with the University of Missouri, Columbia, MO.

Declarations

Conflict of interest

H.S. is CEO of Fascure Therapeutics, LLC, and the Scientific Co-Founder, stockholder, and member of SAB for iTolerance, Inc. E.S.Y. is a consultant for iTolerance. The remaining authors declare no conflict of interest.

Footnotes

Publisher's Note

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

Rakesh Rudraboina and Mohammad Tarique have contributed equally to this work.

Haval Shirwan and Esma S. Yolcu have share senior authorship.

Contributor Information

Haval Shirwan, [email protected].

Esma S. Yolcu, [email protected].

References

1. Siegel RL, et al. Cancer statistics. CA Cancer J Clin. 2023;73(1):17–48. 10.3322/caac.21763. [Abstract] [CrossRef] [Google Scholar]
2. Bray F, et al. Global cancer transitions according to the Human Development Index (2008–2030): a population-based study. Lancet Oncol. 2012;13(8):790–801. 10.1016/S1470-2045(12)70211-5. [Abstract] [CrossRef] [Google Scholar]
3. Morrissey KM, et al. Immunotherapy and novel combinations in oncology: current landscape, challenges, and opportunities. Clin Transl Sci. 2016;9(2):89–104. 10.1111/cts.12391. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
4. O'Donnell JS, Teng MWL, Smyth MJ. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat Rev Clin Oncol. 2019;16(3):151–167. 10.1038/s41571-018-0142-8. [Abstract] [CrossRef] [Google Scholar]
5. Yu L, et al. Opportunities and obstacles of targeted therapy and immunotherapy in small cell lung cancer. J Drug Target. 2021;29(1):1–11. 10.1080/1061186X.2020.1797050. [Abstract] [CrossRef] [Google Scholar]
6. Melero I, et al. NK1.1 cells express 4–1BB (CDw137) costimulatory molecule and are required for tumor immunity elicited by anti-4–1BB monoclonal antibodies. Cell Immunol. 1998;190(2):167–172. 10.1006/cimm.1998.1396. [Abstract] [CrossRef] [Google Scholar]
7. Martin AL, et al. Anti-4-1BB immunotherapy enhances systemic immune effects of radiotherapy to induce B and T cell-dependent anti-tumor immune activation and improve tumor control at unirradiated sites. Cancer Immunol Immunother. 2023;72(6):1445–1460. 10.1007/s00262-022-03325-y. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
8. Sharma RK, Yolcu ES, Shirwan H. SA-4-1BBL as a novel adjuvant for the development of therapeutic cancer vaccines. Expert Rev Vaccines. 2014;13(3):387–398. 10.1586/14760584.2014.880340. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
9. Kamata-Sakurai M, et al. Antibody to CD137 activated by extracellular adenosine triphosphate is tumor selective and broadly effective in vivo without systemic immune activation. Cancer Discov. 2021;11(1):158–175. 10.1158/2159-8290.CD-20-0328. [Abstract] [CrossRef] [Google Scholar]
10. Segal NH, et al. Results from an integrated safety analysis of urelumab, an agonist anti-CD137 monoclonal antibody. Clin Cancer Res. 2017;23(8):1929–1936. 10.1158/1078-0432.CCR-16-1272. [Abstract] [CrossRef] [Google Scholar]
11. Rabu C, et al. Production of recombinant human trimeric CD137L (4–1BBL) Cross-linking is essential to its T cell co-stimulation activity. J Biol Chem. 2005;280(50):41472–41481. 10.1074/jbc.M506881200. [Abstract] [CrossRef] [Google Scholar]
12. Bitra A, et al. Crystal structure of the m4–1BB/4-1BBL complex reveals an unusual dimeric ligand that undergoes structural changes upon 4–1BB receptor binding. J Biol Chem. 2019;294(6):1831–1845. 10.1074/jbc.RA118.006297. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
13. Schabowsky RH, et al. A novel form of 4–1BBL has better immunomodulatory activity than an agonistic anti-4-1BB Ab without Ab-associated severe toxicity. Vaccine. 2009;28(2):512–522. 10.1016/j.vaccine.2009.09.127. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
14. Barsoumian HB, Yolcu ES, Shirwan H. 4–1BB signaling in conventional t cells drives IL-2 production that overcomes CD4+CD25+FOXP3+ t regulatory cell suppression. PLoS ONE. 2016;11(4):e0153088. 10.1371/journal.pone.0153088. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
15. Srivastava AK, et al. SA-4-1BBL and monophosphoryl lipid A constitute an efficacious combination adjuvant for cancer vaccines. Cancer Res. 2014;74(22):6441–6451. 10.1158/0008-5472.CAN-14-1768-A. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
16. Sharma RK, et al. CD4+ T cells play a critical role in the generation of primary and memory antitumor immune responses elicited by SA-4-1BBL and TAA-based vaccines in mouse tumor models. PLoS ONE. 2013;8(9):e73145. 10.1371/journal.pone.0073145. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
17. Madireddi S, et al. SA-4-1BBL costimulation inhibits conversion of conventional CD4+ T cells into CD4+ FoxP3+ T regulatory cells by production of IFN-gamma. PLoS ONE. 2012;7(8):e42459. 10.1371/journal.pone.0042459. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
18. Srivastava AK, et al. Prime-boost vaccination with SA-4-1BBL costimulatory molecule and survivin eradicates lung carcinoma in CD8+ T and NK cell dependent manner. PLoS ONE. 2012;7(11):e48463. 10.1371/journal.pone.0048463. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
19. Sharma RK, et al. SA-4-1BBL as the immunomodulatory component of a HPV-16 E7 protein based vaccine shows robust therapeutic efficacy in a mouse cervical cancer model. Vaccine. 2010;28(36):5794–5802. 10.1016/j.vaccine.2010.06.073. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
20. Sharma RK, et al. 4–1BB ligand as an effective multifunctional immunomodulator and antigen delivery vehicle for the development of therapeutic cancer vaccines. Cancer Res. 2010;70(10):3945–3954. 10.1158/0008-5472.CAN-09-4480. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
21. Sharma RK, et al. Costimulation as a platform for the development of vaccines: a peptide-based vaccine containing a novel form of 4–1BB ligand eradicates established tumors. Cancer Res. 2009;69(10):4319–4326. 10.1158/0008-5472.CAN-08-3141. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
22. Barsoumian HB, et al. A novel form of 4–1BBL prevents cancer development via nonspecific activation of CD4(+) T and natural killer cells. Cancer Res. 2019;79(4):783–794. 10.1158/0008-5472.CAN-18-2401. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
23. Nikitin AY, et al. Classification of proliferative pulmonary lesions of the mouse: recommendations of the mouse models of human cancers consortium. Cancer Res. 2004;64(7):2307–2316. 10.1158/0008-5472.CAN-03-3376. [Abstract] [CrossRef] [Google Scholar]
24. Kim JH, et al. Inhibitory effects of 7-hydroxy-3-methoxy-cadalene on 4-(methylinitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced lung tumorigenesis in A/J mice. Cancer Lett. 2004;213(2):139–145. 10.1016/j.canlet.2004.03.049. [Abstract] [CrossRef] [Google Scholar]
25. Salehinejad J, et al. Immunohistochemical detection of p53 and PCNA in ameloblastoma and adenomatoid odontogenic tumor. J Oral Sci. 2011;53(2):213–217. 10.2334/josnusd.53.213. [Abstract] [CrossRef] [Google Scholar]
26. Patlolla JM, et al. beta-Escin inhibits NNK-induced lung adenocarcinoma and ALDH1A1 and RhoA/Rock expression in A/J mice and growth of H460 human lung cancer cells. Cancer Prev Res (Phila) 2013;6(10):1140–1149. 10.1158/1940-6207.CAPR-13-0216. [Abstract] [CrossRef] [Google Scholar]
27. Galitovskiy V, et al. Development of novel approach to diagnostic imaging of lung cancer with (18)F-Nifene PET/CT using A/J mice treated with NNK. J Cancer Res Ther (Manch) 2013;1(4):128–137. 10.14312/2052-4994.2013-20. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
28. Stabile LP, et al. Syngeneic tobacco carcinogen-induced mouse lung adenocarcinoma model exhibits PD-L1 expression and high tumor mutational burden. JCI Insight. 2021 10.1172/jci.insight.145307. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
29. Narayanapillai SC, et al. Modulation of the PD-1/PD-L1 immune checkpoint axis during inflammation-associated lung tumorigenesis. Carcinogenesis. 2020;41(11):1518–1528. 10.1093/carcin/bgaa059. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
30. Carlino MS, Larkin J, Long GV. Immune checkpoint inhibitors in melanoma. Lancet. 2021;398(10304):1002–1014. 10.1016/S0140-6736(21)01206-X. [Abstract] [CrossRef] [Google Scholar]
31. Tran L, et al. Advances in bladder cancer biology and therapy. Nat Rev Cancer. 2021;21(2):104–121. 10.1038/s41568-020-00313-1. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
32. Diaz-Montero CM, Rini BI, Finke JH. The immunology of renal cell carcinoma. Nat Rev Nephrol. 2020;16(12):721–735. 10.1038/s41581-020-0316-3. [Abstract] [CrossRef] [Google Scholar]
33. Leemans CR, Snijders PJF, Brakenhoff RH. The molecular landscape of head and neck cancer. Nat Rev Cancer. 2018;18(5):269–282. 10.1038/nrc.2018.11. [Abstract] [CrossRef] [Google Scholar]
34. Connors JM, et al. Hodgkin lymphoma. Nat Rev Dis Primers. 2020;6(1):61. 10.1038/s41572-020-0189-6. [Abstract] [CrossRef] [Google Scholar]
35. Gettinger SN, et al. Overall survival and long-term safety of nivolumab (anti-programmed death 1 antibody, BMS-936558, ONO-4538) in patients with previously treated advanced non-small-cell lung cancer. J Clin Oncol. 2015;33(18):2004–2012. 10.1200/JCO.2014.58.3708. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
36. Rizvi NA, et al. Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. Lancet Oncol. 2015;16(3):257–265. 10.1016/S1470-2045(15)70054-9. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
37. Hatic H, Sampat D, Goyal G. Immune checkpoint inhibitors in lymphoma: challenges and opportunities. Ann Transl Med. 2021;9(12):1037. 10.21037/atm-20-6833. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
38. Cao D, et al. Opportunities and challenges in targeted therapy and immunotherapy for pancreatic cancer. Expert Rev Mol Med. 2021;23:e21. 10.1017/erm.2021.26. [Abstract] [CrossRef] [Google Scholar]
39. Onoi K, et al. Immune checkpoint inhibitors for lung cancer treatment: a review. J Clin Med. 2020;9(5):1362. 10.3390/jcm9051362. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
40. Yap TA, et al. Development of immunotherapy combination strategies in cancer. Cancer Discov. 2021;11(6):1368–1397. 10.1158/2159-8290.CD-20-1209. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
41. Yu WD, et al. Mechanisms and therapeutic potentials of cancer immunotherapy in combination with radiotherapy and/or chemotherapy. Cancer Lett. 2019;452:66–70. 10.1016/j.canlet.2019.02.048. [Abstract] [CrossRef] [Google Scholar]
42. Gotwals P, et al. Prospects for combining targeted and conventional cancer therapy with immunotherapy. Nat Rev Cancer. 2017;17(5):286–301. 10.1038/nrc.2017.17. [Abstract] [CrossRef] [Google Scholar]
43. Chester C, et al. Immunotherapy targeting 4–1BB: mechanistic rationale, clinical results, and future strategies. Blood. 2018;131(1):49–57. 10.1182/blood-2017-06-741041. [Abstract] [CrossRef] [Google Scholar]
44. Etxeberria I, et al. New emerging targets in cancer immunotherapy: CD137/4-1BB costimulatory axis. ESMO Open. 2020;4(Suppl 3):e000733. [Europe PMC free article] [Abstract] [Google Scholar]
45. Sharma RK, et al. Tumor cells engineered to codisplay on their surface 4–1BBL and LIGHT costimulatory proteins as a novel vaccine approach for cancer immunotherapy. Cancer Gene Ther. 2010;17(10):730–741. 10.1038/cgt.2010.29. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]

Articles from Cancer Immunology, Immunotherapy : CII are provided here courtesy of Springer

Citations & impact 


Impact metrics

Jump to Citations

Alternative metrics

Altmetric item for https://www.altmetric.com/details/157179246
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/157179246

Article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Funding 


Funders who supported this work.

U.S. Department of Defense (1)