Movatterモバイル変換


[0]ホーム

URL:


Skip to main content
                                  NCBI home page
Search in PMCSearch
As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsement of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more:PMC Disclaimer | PMC Copyright Notice
NIHPA Author Manuscripts logo
. Author manuscript; available in PMC: 2020 Oct 12.

Radiation-Induced Enhancement of Antitumor T-cell Immunity by VEGF-Targeted 4–1BB Costimulation

Brett Schrand1,Bhavna Verma1,Agata Levay1,Shradha Patel1,Iris Castro1,Ana Paula Benaduce2,Randall Brenneman2,Oliver Umland3,Hideo Yagita4,Eli Gilboa1,Adrian Ishkanian2
1Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida,
2Department of Radiation Oncology, Dodson Interdisciplinary Immunotherapy Institute, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida.
3Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida,
4Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan.

Authors’ Contributions

Conception and design: B. Schrand, B. Verma, R. Brenneman, A. Ishkanian Development of methodology: B. Schrand, I. Castro, R. Brenneman, O. Umland, A. Ishkanian

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): B. Schrand, B. Verma, S. Patel, A.P. Benaduce, A. Ishkanian

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): B. Schrand, B. Verma, S. Patel, I. Castro, R. Brenneman, O. Umland, A. Ishkanian

Writing, review, and/or revision of the manuscript: B. Schrand, S. Patel, H. Yagita, E. Gilboa, A. Ishkanian

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A. Levay, S. Patel, H. Yagita, A. Ishkanian Study supervision: E. Gilboa, A. Ishkanian

Corresponding Author: Adrian Ishkanian, University of Miami School of Medicine, 1550 NW 10th Avenue, Suite 110, Miami, FL 33136. Phone: 305-243-5454; Fax: 305-243-1854;aishkanian@med.miami.edu

Issue date 2017 Mar 15.

PMCID: PMC7549621  NIHMSID: NIHMS1627889  PMID:28082399
The publisher's version of this article is available atCancer Res

Abstract

Radiotherapy can elicit systemic immune control of local tumors and distant nonirradiated tumor lesions, known as the abscopal effect. Although this effect is enhanced using checkpoint blockade or costimulatory antibodies, objective responses remain suboptimal. As radiotherapy can induce secretion of VEGF and other stress products in the tumor microenvironment, we hypothesized that targeting immunomodulatory drugs to such products will not only reduce toxicity but also broaden the scope of tumor-targeted immunotherapy. Using an oligonucleotide aptamer platform, we show that radiation-induced VEGF-targeted 4–1BB costimulation potentiated both local tumor control and abscopal responses with equal or greater efficiency than 4–1BB, CTLA-4, or PD1 antibodies alone. Although 4–1BB and CTLA-4 antibodies elicited organ-wide inflammatory responses and tissue damage, VEGF-targeted 4–1BB costimulation produced no observable toxicity. These findings suggest that radiation-induced tumor-targeted immunotherapy can improve the therapeutic index and extend the reach of immunomodulatory agents.

Introduction

Although the main function of both radiotherapy and chemotherapy is cytotoxic tumor control, both modalities can also elicit protective antitumor immune responses through “immunogenic cell death” that can be further enhanced with immune-potentiating strategies (1,2). Albeit rarely, radiotherapy can also lead to the regression of distant nonirradiated tumor lesions after local tumor ablative therapy. Although the mechanism of this “abscopal effect” is not fully elucidated, it is most likely mediated through a systemic immune response that is capable of controlling the growth of the nonirradiated tumor lesions.

Preclinical studies in mice have demonstrated the immune-potentiating nature of local tumor radiation (24), and therefore could serve as an alternative to classical vaccination (RadVax). In mice, radiation-induced antitumor immunity can be further enhanced using immune-stimulatory treatments, such as checkpoint blockade with CTLA-4 or PD-1 antibodies, costimulation with 4–1BB, CD40, LIGHT antibodies or their ligands, treatment with cytokines such as Flt-3 or IFNα, or blocking TGFβ action (35). Importantly, combination with immune therapy enhanced the abscopal effect, the control of contralaterally implanted or metastatic lesions, although the effect was more modest (611). In case reports and early clinical trials combining local radiation with systemically delivered nontargeted immunomodulatory agents demonstrated some modest but encouraging local and abscopal responses (3,4,12).

Mirroring preclinical studies in mice, treatment of patients with immunomodulatory antibodies such as blocking anti-CTLA-4 or agonistic 4–1BB antibodies, and to a lesser extent PD-1 or PD-L1 antibodies, can elicit significant autoimmune pathologies (13,14). Cotreatment with anti-CTLA-4 and anti-PD-1 antibodies was accompanied by significant toxicities above what was seen with anti-CTLA-4 and anti-PD-1 antibody alone (15), and coadministration of CTLA-4 antibodies and the BRAF inhibitor vemurafenib elicited unacceptable toxicities (16). It is, therefore, reasonable to expect that toxicities will escalate with the development of increasingly potent immune therapies, including combinations of radiation and immune-potentiating therapies. Eliciting a systemic immune response devoid of serious adverse effects that is capable of controlling distant nontreated metastatic lesions remains a main challenge in developing an effective radiation-induced immune strategy.

4–1BB is a case in point. 4–1BB is a major immune-stimulatory receptor expressed on activated CD8+ T cells (17). Systemic administration of agonistic 4–1BB antibodies to mice potentiates antitumor immunity, but also elicits organ-wide inflammatory responses and liver damage (18,19), and severe liver toxicity in cancer patients (20). Drug toxicity can be reduced by targeting the immune-modulatory agents to the tumor lesion. We have shown that 4–1BB costimulation can be targeted to tumors by conjugating an immune-stimulatory 4-lBB-binding oligonucleotide aptamer to aptamers that bind to products that are upregulated in the tumor stroma like VEGF or osteopontin (21). The cell-free chemically synthesized nuclease-resistant oligonucleotide aptamers represent a novel and emerging platform for generating ligands with desired specificity that offer potential advantages in term of development, manufacture, cost, and reduced or lack of immunogenicity (22). Underscoring the potency and broad applicability of tumor stroma-targeted 4–1BB costimulation, in preclinical murine tumor models systemic administration of a VEGF-targeted 4–1BB aptamer conjugate engendered potent antitumor immunity against multiple tumors of distinct origin that were substantially more pronounced than what has been previously reported. Importantly, the tumor-targeted 4–1BB aptamer ligand exhibited a significantly superior therapeutic index, ratio of efficacy to toxicity, compared with nontargeted administration of an agonistic 4–1BB antibody (21). Thus, targeting 4–1BB costimulation to the irradiated tumor as well as to distant tumor lesions will ameliorate the known toxicities of 4–1BB antibodies that could be exacerbated upon combination with irradiation.

VEGF is a key angiogenic factor expressed in virtually all tumors of every type of cancer analyzed (23,24), suggesting that VEGF-targeted immune modulation will be applicable to a broad range of tumors of distinct origin. In mice, VEGF-4–1BB aptamer conjugates were effective against melanoma, breast cancer, fibrosarcoma, and glioma (21). Yet, in human patients, the levels of VEGF vary widely from lesion to lesion and patient to patient, correlating with the histologic grade of the tumor, metastatic state, clinical stage, and outcome (2529). Consequently, VEGF-targeted immune modulation will not be as broadly applicable and effective as suggested by the murine studies (21). Here we describe a novel method to extend tumor-targeted immunotherapy also to VEGFlow tumors using radiation. Radiation causes tissue injury triggering an angiogenic process akin to wound healing (24,30,31) that is accompanied by upregulation of VEGF expression, especially in lesions expressing low levels of VEGF. This was seen in syngeneic murine tumors (refs.32,33 and see below) xeno-grafted human tumors (34), and importantly in human tumorsin situ (3537). We, therefore, hypothesized that VEGF-targeted 4–1BB costimulation will not only potentiate radiation-induced antitumor immunity and reduce the toxicities associated with systemic administration of 4–1BB ligands, but will also broaden the scope of VEGF-targeted immune modulation to those tumor lesions that otherwise express low levels of VEGF.

Materials and Methods

Construction of aptamer conjugates

A 2′-fluoro-pyrimidine-modified dimeric 4–1BB RNA aptamer transcribedin vitro from a DNA template described (21) extended at the 3′ end with a linker sequence 5′-UCCCG-CUAUAAGUGUGCAUGAGAAC-3 was annealed to a chemically synthesized (IDT) VEGF aptamer (38) via a complementary linker sequence engineered at the 3′ end. Equimolar amounts of 4–1BB and VEGF aptamer was mixed, heated to 82°C, and cooled to room temperature. Annealing efficiency, monitored by agarose gel electrophoresis, was >90%. To preventin vivo annealing of the two aptamers, they were annealed separately with a 2-fold excess of complementary linker sequence before tail vein injection.

Tumor immunotherapy studies

The facilities at the University of Miami’s Division of Veterinary Resources are fully accredited by the Association for Assessment and Accreditation of Laboratory Animal Care and U.S. Drug Administration. An Office of Laboratory of Animal Welfare assurance is on file, ensuring that humane animal care and use practices, as outlined in the Guide for the Care and Use of Laboratory Animals, are followed. Of note, 5- to 6-week-old and Balb/c (H-2d)mice were purchased from The Jackson Laboratory and used within 1 to 3 weeks.

Subcutaneous tumor models

Six- to 8-week-old Balb/c mice were injected subcutaneously on the right flanks with 1.0 × 105 4T07 or CT26 tumors cells. Tumors were allowed to grow to about 75 to 100 mm3, at which point mice were anesthetized with a mixture of ketamine and xylazine (80–100 and 10 mg/kg, respectively) by intraperitoneal injection using an insulin syringe. Once anesthetized, animals were placed in a custom lead shielding apparatus to expose the tumor but minimally expose the rest of the animal. The apparatus was placed into and RS-2000 irradiator (RadSource) with 225 kV energy at an approximate rate of 1.8 Gy/minute where tumors were irradiated with a single fraction of 12 Gy. At days 6, 8, and 10 post irradiation, animals were administered with 100 pmol of aptamer conjugate or mixture of via tail vein, or with 800 pmol of 4–1BB (clone 3H3, BioXcell).

3-Methylcholanthrene-induced fibrosarcoma model

Balb/c mice were injected subcutaneously with 400 μg of 3-methylcholanthrine (MCA) in castor oil. When tumors reached an approximate size of 100 mm3, mice were irradiated as previously described with 20 Gy. As indicated, mice were injected with 100 pmol of aptamer conjugate or mixture of via tail vein three times, 3 days apart. Mice were sacrificed when tumor exceeded 1,200 mm3 or mice exhibited signs of morbidity.

Subcutaneous abscopal model

Balb/c mice were implanted subcutaneously on the right flank with 2.0 × 104 4T1 cells. Eight days later, when right flank tumor started to become palpable, 2.0 × 104 4T1 cells were injected subcutaneously on the left flank. At day 13 post right flank implantation, when tumors reached 75 to 100 mm3, the mice were anesthetized and irradiated as described above followed by 100 pmol administration of aptamer conjugate via tail vein injection on days 3, 6, and 9 postirradiation. Alternatively, 200 μg of CTLA-4 (clone 9H10, BioXcell) and/or 200 μg PD-1 antibody (clone RMP1–14, produced by HY) were injected intraperitoneally.

Metastatic abscopal model

Balb/c mice were implanted subcutaneously on the right flank with 2.0 × 104 4T1 cells. Eight days later, when right flank tumor started to become palpable, 2.0 × 104 4T1 cells were injected intravenously. At day 13 after right flank implantation, when tumors were roughly 75 to 100 mm3, the mice were anesthetized and irradiated as described above with a single dose of 12 Gy followed by 100 pmol administration of aptamer conjugate via tail vein injection on days 3, 6, and 9 post irradiation. At day 21 post intravenous injection, when untreated mice started to show signs of morbidity, mice were sacrificed and the weight of lungs was determined as a measure of tumor burden.

Toxicity studies

Balb/c mice bearing 4T07 tumors were irradiated and treated with 100 pmol of aptamer conjugate, 100 or 800 pmol of 4–1BB 3H3 agonistic antibody, or with 800 pmol of isotype. Mice were sacrificed 5 days after the last treatment. Lung, liver, and spleen weights were weighed, fixed in formalin, and embedded followed by staining with hematoxylin and eosin or homogenized to generate a single-cell suspension and CD8+ T cells were stained with fluorophore-labeled antibody for flow cytometry.

FACS analysis

Cells were incubated with fixable viability dye eFluor780 for 10 minutes at 4°C and washed twice in FACS buffer prior to staining in order distinguish live cells from the dead cells. Cells were incubated with Fc-blocking antibodies for 10 minutes at 4°C, then incubated for 30 minutes at 4°C with CD3 FITC, CD4 PerCP Cy5.5, CD25 PE-Cy7, CD62LeFluor450, FOXP3 PE, KLRG1APC, CD127 AF700, CD11B PerCp Cy5.5, CD11cAPC, GR1 PE, F4/80 PE-Cy7 from eBioscience, and CD8 BV605 and CD44 BV510 from Biolegend. For intracellular staining, cells were fixed and permeabilized with FOXp3 Staining Buffer Kit (eBioscience) according to manufacturer’s instructions. Cells were incubated for 30 minutes at room temperature in dark with Foxp3-PE and analyzed using the CytoFLex (BD Biosciences) and Kaluza software (Beckman Coulter).

Isolation of tumor-infiltrating T cells

Tumors were harvested 2 days after last treatment and weighed. Cells were isolated by dissecting tumor tissue into small fragments followed by digesting in 1 mg/mL of collagenase in complete RPMI media (10% FBS, 1% penicillin, streptomycin) prior to using Gentle MACS Dissociator. Cell suspension was passed through a 70 μm nylon strainer to obtain a single-cell population. Cells were washed twice with FACS buffer and stained as described above and analyzed by flow cytometry.

Cytokine analysis in the serum

Plasma levels of IFNγ, GM-CSF, TNFα, MCP-1, ILlα, ILlβ, IL10, and IL6 were analyzed by Multianalyte Flow Assay Kit (BioLegend) according to the manufacturer’s instructions. Mean fluorescent intensity (MFI) was plotted for each analyte in the serum sample. Data collection and analysis was done using CytoFlex (BD Biosciences).

Tumor homing

32P-labeled VEGF aptamer was generated by T4 kinase end labeling using γP32 ATP and purified using G25 Microspin Columns (GE). A total of 2 × 105 cpm of material was injected via tail vein into mice bearing both an irradiated and nonirradiated subcutaneous 4T07 tumor that were approximately 150 mm3. Forty-eight hours postinjection, mice were sacrificed, tumors were excised, placed in 500 μL of scintillation fluid, and counted with a Scintillation counter.

1HC of VEGF in tumor tissue

4T1, 4T07, and CT26 subcutaneously established tumors were resected and embedded in paraffin. Nonspecific immunoreactivity in slide-mounted tissue sections was blocked with Serum-Blocking Reagent D (R&D Systems), and incubated with goat polyclonal anti-mouse VEGF at 1:20 (R&D Systems) at 4°C overnight, washed with PBS, and incubated with biotinylated anti-goat secondary antibody (R&D Systems) for 40 minutes, followed by high-sensitivity streptavidin (HSS)-horseradish peroxidase (HRP; R&D Systems) for 30 minutes. Slides were washed with PBS and then incubated with an HRP-reactive DAB chromogen (R&D Systems) for 7 minutes. Slides were rinsed in H2O, incubated in hematoxylin (Vector) at 1:5 for 3 minutes and incubated with 0.1% sodium bicarbonate as a bluing agent, at 1:5 for 3 minutes, followed by rinses with H2O. Slides were dehydrated in increasing concentrations of alcohol (70%, 90%, 100%; 3 minutes each), briefly washed in xylene, and cover-slipped using mounting medium (Richard-Allan Scientific). Negative control slides for VEGF IHC were prepared by completing the above protocol in the absence of primary antibody.

Hematoxylin and eosin staining

Lungs, liver, and small intestines were fixed in 10% formalin overnight at room temperature and embedded in paraffin. Slides were stained with eosin (Across organics) for 5 minutes, followed by 10 minutes of hematoxylin staining (Vector), incubated with 0.1% sodium bicarbonate as a bluing agent, rinsed in H2O, and then dehydrated in increasing concentrations of alcohol, as described earlier.

Intracellular granzyme B staining

CD8+ T cells were purified from splenocytes of treated animals using a CD8 Enrichment Kit (Miltenyi Biotec). Cells were incubated overnight in the presence of murine CD3 antibody. Eighteen hours later, cells were treated with Fix/Perm (Becton Dickinson) and stained with an antimurine granzyme B antibody (Becton Dickinson).

Results

Radiation sensitizes VEGFlow-expressing tumors to VEGF-targeted 4–1BB costimulation

We have previously shown that VEGF-targeted 4–1BB costimulation using a systemically administered VEGF-4–1BB aptamer conjugate enhanced tumor immunity in multiple tumor models including the VEGF-expressing 4T1 breast carcinoma tumors. VEGF-targeted 4–1BB costimulation was, however, less effective against 4T07 tumors, a nonmetastatic sibling of 4T1 tumors that expresses low levels of VEGF (21). Given that radiation upregulates intratumoral VEGF expression, we tested the hypothesis that local tumor radiation will enhance the susceptibility of VEGFlow 4T07 tumors to VEGF-targeted 4–1BB costimulation. Using immunohistochemical analysis,Fig. 1A shows that radiation of subcutaneously implanted 4T07 tumors upregulates VEGF expression to levels comparable with that of 4T1 tumors. There was no detectable increase in VEGF levels in the circulation of 4T1 tumor-bearing mice that were or were not irradiated (data not shown). Using mice bearing two subcutaneous 4T07 tumors implanted at opposite flanks,Fig. 1B shows that intravenously administered VEGF, but not control, aptamer accumulate in the irradiated, but not in the nonirradiated, tumor lesion. The VEGF-4–1BB aptamer conjugate accumulated preferentially in irradiated or nonirradiated 4T1 tumors compared with normal organs (Supplementary Fig. S1).

Figure 1.

Figure 1.

Radiation potentiates VEGF-4–1BB aptamer conjugate-mediated antitumor immunity in the VEGFlow 4T07 tumor model.A, VEGF expression in 4T1 and 4T07 tumors. 4T1 or 4T07 tumor cells were injected subcutaneously into Balb/c mice and when tumors reached about 150 mm3, they were resected and stained for VEGF expression as described in Materials and Methods.B, Homing of VEGF aptamer to VEGF-expressing tumors. Balb/c mice were implanted with 4T07 tumors in the opposite flanks. When tumors reached an average diameter of 75 mm3, the tumors at the right flank were irradiated (XRT) with either 2 or 20 Gy and 7 days later32P-labeled VEGF (VEGF) or scrambled aptamer were administered via the tail vein. Twenty-four hours later mice were sacrificed, tumors excised, and radioactivity was determined by scintillation counting. The difference between the 2 and 20 Gy irradiated groups injected with the VEGF conjugate and any of the other groups was statistically significant (P < 0.01) C, 4T07 tumor cells were implanted subcutaneously in Balb/c mice and irradiated when tumor reached an average of 75 mm3. Seven days later, mice were treated with VEGF-4–1BB conjugate or with a mixture of VEGF and 4–1BB aptamers administered via the tail vein three times 2 days apart starting 6 days postirradiation.D, 4T07 tumor cells were implanted subcutaneously in Balb/c mice and irradiated when tumor reached an average of 75 mm3. Six days later, mice were treated with 100 pmol (1×) of VEGF-4–1BB conjugate, mixture of VEGF and 4–1BB aptamers, or with 800 pmol (8×) of an agonistic 4–1BB antibody or isotype control (IgG) three times 2 days apart. Aptamer was administered via the tail vein, whereas antibody was administered intraperitoneally.

To determine whether radiation-induced tumor accumulation of VEGF-4–1BB aptamer conjugate leads to control of tumor growth mice were treated systemically with the VEGF-4–1BB conjugates by tail vein injection. As shown inFig. 1C, mono-therapy with either radiation or VEGF-4–1BB conjugate had a modest effect on the rate of tumor growth but no appreciable impact on survival, whereas a combination of radiation and VEGF-4–1BB conjugate significantly inhibited tumor growth leading to the long-term survival of 50% of the treated mice. Consistent with the targeted nature of the action of VEGF-4–1BB aptamer conjugates, coadministration of unconjugated VEGF and 4–1BB aptamers did not potentiate the radiation effects on tumor growth, showing that the synergy between radiation and VEGF-4–1BB conjugate was not a result of the separate contributions of VEGF and/or 4–1BB aptamers.

We next compared the antitumor activity of the VEGF-targeted 4–1BB aptamer conjugate to that of an agonistic 4–1BB antibody when used in combination with irradiation. As shown inFig. 1D, both antibody and aptamer conjugate potentiated the ability of radiation to inhibit tumor growth, leading to the long-term survival of 50% to 60% of the treated mice in each group. On a molar basis, however, 8-fold more antibody was required to achieve a comparable antitumor effect. VEGF-targeted 4–1BB costimulation was also able to potentiate radiation-induced tumor immunity in the autochthonous MCA-induced fibrosarcoma model. As shown inSupplementary Fig. S2, combined radiation and VEGF-4–1BB conjugate treatment significantly enhanced the overall survival of the treated group, with 2 out of 6 mice exhibiting complete tumor regression, whereas either radiation orVEGF-4-1BB conjugate administration alone delayed tumor growth and enhanced survival though all mice eventually succumbed to the tumor and were sacrificed.

Therapeutic dose of 4–1BB antibody, but not VEGF-4–1BB aptamer conjugate, elicits nonspecific immune activation and inflammation

Therapeutic doses of 4–1BB antibody (18,19), but not VEGF-4–1BB conjugates elicited immune pathologies in mice (21), that were seen also in patients (20). Because combination therapies that lead to enhanced antitumor immune responses could exacerbate such autoimmune sequelae, we evaluated whether cotherapy of radiation and 4–1BB costimulation will induce and/or exacerbate immune pathology. In murine tumor studies, 4–1BB antibodies were shown to synergize with radiation but toxicities were not evaluated (3941). In this study, the livers and spleens of mice treated with a therapeutic dose of the agonistic 4–1BB antibody (8×) shown inFig. 1D were enlarged (Fig. 2A andB), the proportion of CD8+ T cells in both organs was elevated (Fig. 2C andD), IFNγ levels were increased in the serum (Fig. 2E), and the splenic CD8+ T cells exhibited an activated-effector phenotype expressing granzyme B (Fig. 2F). In contrast, mice treated with a therapeutic dose ofVEGF-4-1BB aptamer conjugate (1×) did not exhibit changes in organ weight, CD8+ T-cell percentage, or activation status. It is noteworthy that even 8-fold less, subtherapeutic doses of antibody (1×) elicited measurable immune anomalies, increased liver and spleen weight (Fig. 2A andB), increased proportion of CD8+ T cells in the spleen (Fig. 2C), and a slight increase in granzyme B-expressing cells (Fig. 2F). Consistent with this, treatment with radiation and 4–1BB antibody, but not VEGF-4–1BB aptamer conjugate, led to significant increases in TNF, MCP-1, and IFNγ in the circulation, whereas increases in IL1α, IL1β, IL10, and IL6 exhibited a trend that did not reach statistical significance (Supplementary Fig. S3). Treatment of the tumor irradiated mice with the therapeutic dose of 4–1BB antibody (8×), but not VEGF-4–1BB (1×), was accompanied by a massive infiltration of leukocytes in the lung (Fig. 2G) and elicited characteristic inflammatory foci, local accumulation of leukocytes, in the livers of the treated mice (Fig. 2H). Hepatic toxicity was the major immune-related toxicity in patients treated with a 4–1BB antibody (20). Inflammatory foci could be also detected in mice treated with 8-fold lower subtherapeutic doses of antibody (arrows,Fig. 2H). Acute phase proteins alanine transaminase (ALT) and aspartate transaminase (AST) levels in the circulation, a measure of liver dysfunction, were elevated in tumor-bearing mice treated with radiation and 4–1BB antibody compared with that of mice treated with radiation and VEGF-4–1BB aptamer conjugates (Fig. 2I). Note that radiation and/or tumor alone increased AST and ALT levels, whereas cotreatment with VEGF-4–1BB aptamer conjugate prevented or reduced the increase, probably reflecting the reduced tumor volume in the aptamer conjugate-treated mice. In contrast, although cotreatment with 4–1BB antibody also led to a comparable decrease in tumor volume (Fig. 1D), the AST and ALT levels remained high. Overall, these experiments suggest that in combination with radiation, VEGF-targeted 4–1BB costimulation exhibit a more than 8-fold higher therapeutic index then the 4–1BB antibodies.

Figure 2.

Figure 2.

Combination of radiation with 4-1BB antibody, but not with VEGF-4–1BB aptamer conjugate, elicits nonspecific immune responses and inflammation. Balb/c mice were injected subcutaneously with 4T07 tumor cells and when they reached an average diameter of 75 mm3, tumors were irradiated. One hundred pmol (1×) of VEGF-4–1BB aptamer conjugate or 800 pmol (8×) of either 4–1BB antibody or IgG isotype control was administered 6 days later via the tail vein three times 2 days apart. Mice were sacrificed 5 days following the last administration. Liver(A) and spleen(B) weight. Percentage of CD8+ cells in the liver(C) and spleen(D). E andF, Serum IFNγ(E) and percentage of intracellular granzyme B-expressing CD8+ cells(F). Tissue sections from the lung(G) and liver(H) were stained with hematoxylin and eosin and visualized by light microscopy. Arrows, H&E staining inflammatory foci in the liver treated with 100 pmol (1×) of 4–1BB antibody.I, ALT and AST levels in the circulation 14 days after irradiation (3 mice/group). NS, nonsignificant.

VEGF-targeted costimulation enhances radiation-induced abscopal effect

To determine whether VEGF-targeted 4–1BB costimulation with aptamer conjugates can potentiate the abscopal effect of radiation, 4T1 tumor cells were implanted subcutaneously in the right flank of a mouse and 8 days later 4T1 tumor cells were implanted also in the left flank. When tumors in the right flank reached about 75–100 mm3, they were irradiated and mice were treated systemically with either VEGF-4–1BB aptamer conjugates, CTLA-4 antibody, PD-1 antibody, or a combination of PD-1 and CTLA-4 antibodies. CTLA-4 and PD-1 antibodies were previously shown to induce abscopal effects in mice that underwent local tumor radiation (8,10). As shown inFig. 3A (tumor size at day 19 post irradiation, representative of other time points) andSupplementary Fig. S4A andS4B, time course of tumor growth), treatment of tumors by local radiation or systemic administration of antibodies or aptamer conjugate inhibited the growth of the irradiated tumor to a comparable extent, and combined radiation with either treatment was significantly enhanced.

Figure 3.

Figure 3.

Inhibition of irradiated and nonirradiated tumors in mice treated with PD-1 antibody, CTLA-4 antibody, and VEGF-4–1BB aptamer conjugates. 4T1 tumor cells were implanted subcutaneously in the right and left flanks of Balb/c mice at days 0 and 8, respectively. At day 13 when the tumors in the right flank reached an average size of 75 mm3, they were irradiated once with 12 Gy, and 3 days later treated as shown three times 2 days apart. Tumor volume at day 19 postirradiation is shown for the irradiated tumor, right flank (RF;A) and for nonirradiated tumor, left flank (LF; seven or eight mice per group;B). Time course of tumor growth is shown inSupplementary Fig. S4. Statistical analysis:A, differences between untreated group and groups treated by radiation, CTLA-4 antibody, PD-1 antibody, CTLA-4 + PD-1 antibody, or VEGF-4–1BB conjugate were significant (P < 0.05), whereas no significant differences were seen among the treated groups. Combination with radiation was significant for all treatments, PD-1,P = 0.0075; CTLA-4,P = 0.0191; PD-1 + CTLA-4,P = 0.0164; VEGF-4–1BB,P = 0.0071.B, Differences between untreated or radiated group and groups treated with CTLA-4 antibody, PD-1 antibody, CTLA-4 + PD-1 antibody, and VEGF-4–1BB conjugate were significant (P < 0.05). Differences between groups treated with radiation and PD-1 antibody or with PD-1 + CTLA-4 antibody and groups treated with antibody alone was not significant. Differences were significant between CTLA-4 antibody versus radiation + CTLA-4 antibody,P = 0.0265, and VEGF-4–1BB aptamer conjugate and radiation + VEGF-4–1BB aptamer conjugate groups,P = 0.0412.C, Survival of mice treated as described above. Mice were sacrificed when either tumor reached 1,000 mm3. Difference between untreated group and either radiation, PD-1 antibody, radiation + PD-1 antibody, CTLA-4 antibody, and VEGF-4–1BB aptamer conjugate-treated groups showed a trend but did not reach statistical significance. Differences between radiation + CTLA-4 antibody, PD-1 + CTLA-4 antibodies with or without radiation, and radiation + VEGF-4–1BB aptamer conjugate-treated groups and rest of the treatment groups was significant. Differences between radiation + VEGF-4–1BB aptamer conjugates or with PD-1 + CTLA-4 antibodies groups and PD-1 + CTLA-4 antibodies or radiation + CTLA-4 antibody-treated groups exhibited a trend but did not reach statistical significance.

The effects of the treatment combinations on the nonirradiated tumor are shown inFig. 3B andSupplementary Fig. S4C andS4D. Radiation alone had no effect, underscoring the challenging nature of inducing an abscopal effect, whereas treatment with antibodies or aptamer conjugate without irradiation reduced tumor growth to a comparable extent. When combined with radiation, CTLA-4 antibody and VEGF-4–1BB aptamer conjugate treatment significantly enhanced inhibition of tumor growth, whereas PD-1 antibody did not. Somewhat unexpectedly, whereas combination of CTLA-4 + PD-1 antibodies enhanced radiation effect on the irradiated tumor, as was previously reported except for using PD-L1 antibody and a different mouse tumor model (42), the abscopal effect was not enhanced. The reason for that is currently under investigation. Measuring prolongation of survival (Fig. 3C), which accounts for the combined contribution of both irradiated and nonirradiated tumor, radiation combined with PD-1 + CTLA-4 antibodies or with VEGF-4–1BB aptamer conjugates were most effective. Radiation with CTLA-4 antibody, or PD-1+ CTLA-4 antibody without irradiation were also effective but less so, while the remaining treatment groups showed a trend toward extended survival that did not reach statistical significance.

To assess the abscopal effect on lung metastasis, 4T1 tumor cells were administered intravenously at day 5 prior to radiation of the subcutaneously implanted tumors. Experiment was terminated when tumor-bearing mice in the untreated group had to be sacrificed. Lung metastasis was determined measuring lung weight. As shown inFig. 4C, radiation of the subcutaneous tumors followed by systemic therapy with the VEGF-4–1BB conjugate significantly inhibited lung metastasis, whereas radiation alone or radiation coadministered with unconjugated VEGF and 4–1BB aptamers had a modest effect. Note that radiation alone had a less pronounced effect on the nonirradiated lung métastasés (Fig 4C) compared with that of the irradiated tumor (Fig. 4A andB), underscoring the challenging nature eliciting an effective abscopal effect.

Figure 4.

Figure 4.

Inhibition of nonirradiated lung metastasis by local radiation of subcutaneously implanted tumors and treatment with VEGF-4–1BB aptamer conjugates. 4T1 tumor cells were implanted subcutaneously in the right flank of Balb/c mice, and 8 days later mice were injected intravenously with 4T1 cells. Five days after intravenous injections when the subcutaneously growing tumors reached an average size of 75 mm3, they were irradiated with 12 Gy and 3 days later treated with VEGF-4–1BB aptamer conjugates or with a mixture of VEGF and 4–1BB aptamers three times 2 days apart. Five days before irradiation, 4T1 tumor cells were injected intravenously to establish métastasés in the lung. Mice were sacrificed when mice in the control group showed signs of morbidity (21 days post-intravenous tumor administration) and tumor burden was determined.A, Volume of subcutaneously implanted tumor at day of sacrifice.B, Time course of subcutaneous tumor growth.C, Lung weights measured when mice were sacrificed.

Combination of radiation and CTLA-4 antibody, but not VEGF-4–1BB aptamer conjugate, elicits organ-wide inflammation

In murine studies, treatment with CTLA-4 antibodies was shown to enhance radiation-induced antitumor immunity but toxicities were not evaluated (7,10,42). In human patients, treatment with CTLA-4 antibody (Ipilumimab) was accompanied by immune-related adverse effects (irAE) that are generally mild and reversible but occasionally can be severe and life threatening (13,14). Enterocolitis was the most frequent toxicity defined by grade 3/4 clinical presentation and/or biopsy documentation presents with diarrhea, pain, and sometimes bowel perforation requiring surgical intervention and can be lethal (43).

To determine whether CTLA-4 antibody or VEGF-4–1BB aptamer conjugate treatment in combination with radiation elicit irAEs in mice, we analyzed the small intestine, liver, and lungs of mice for inflammatory infiltrates. Recapitulating the scenario in human patients, immunohistochemical analysis of small intestine of mice treated with CTLA-4 antibody alone or in combination with radiation and/or PD-1 antibody exhibited a significant increase in leukocytic infiltrates and deformation of the crypt architecture (Fig. 5;Supplementary Fig. S5). Strikingly, radiation and/or VEGF-4–1BB aptamer conjugate showed no sign of increased leukocytic infiltrates or tissue damage. Similar pattern of inflammatory infiltrates was seen in the livers and lungs (Supplementary Figs. S6 andS7). Radiation in combination with CTLA-4 + PD-1 antibodies, and to a lesser combination with CTLA-4 antibody alone, but not with VEGF-4–1BB aptamer conjugate, elicited significant increases of MCP-1, IL6, and GM-CSF in the circulation, whereas radiation in combination with VEGF-4–1BB aptamer conjugates did not (Supplementary Fig. S8). VEGF-targeted 4–1BB costimulation, therefore, exhibited a significantly increased therapeutic index compared with CTLA-4 antibody or CTLA-4 and PD-1 antibody combinations, with or without radiation.

Figure 5.

Figure 5.

Hematoxylin and eosin staining of sections from small intestine.A, Magnification, ×10;B, Magnification, ×40. Mice were treated as described inFig. 2. Arrows, increased lymphocytic infiltrates in mice treated with CTLA-4 antibody, CTLA-4 + PD-1 antibodies with or without radiation, compared with untreated mice or mice treated with VEGF-4–1BB aptamer conjugate with or without radiation. Contours indicate areas of dense leukocytic infiltrates that appear to have deformed or displaced the normal crypt architecture seen in mice treated with CTLA-4 or PD-1 + CTLA-4 antibodies with or without radiation (see alsoSupplementary Fig. S5).

Treatment with VEGF-4–1BB aptamer conjugates enhances the infiltration of immune cells into irradiated tumors

To investigate the immunologic basis for the antitumor activity of radiation and VEGF-4–1BB aptamer conjugates, we evaluated the nature of tumor-infiltrating lymphocytes (TIL). As shown inFig. 6, radiation promotes the infiltrations of a diverse subsets of immune cells, including CD4+ and CD8+T cells, Ml-polarized macrophages, and B cells that was significantly enhanced by cotreatment with VEGF-4–1BB aptamer conjugates, whereas treatment with VEGF-4–1BB aptamer conjugate alone had a minimal effect at best. Conversely, radiation + VEGF-4–1BB aptamer conjugate treatment led to a reduction in tumor-infiltrating Treg and increase in the ratio of CD8+ T cells/foxp3+ Treg that has been associated with tumor immunogenicity (44). The immune-potentiating effects of cotreatment with VEGF-4–1BB aptamer conjugate was also indicated by a significant increase in overall cell death at the tumor site over that of radiation alone (Fig. 6). These findings are consistent with the immune-promoting properties of local tumor irradiation (24) and the enhanced antitumor activity of combination with VEGF-targeted 4–1BB costimulation (Figs. 1,3,4;Supplementary Figs. S3 andS4).

Figure 6.

Figure 6.

Tumor infiltration of immune cell subsets. Subcutaneously implanted 75 to 100 mm3 4T1 tumor-bearing mice were irradiated and/or treated with VEGF-4–1BB aptamer conjugates 3 days postirradiation, three times 2 days apart, and 2 days after the last treatment tumors were excised, homogenized, and analyzed for immune cell subsets by flow cytometry or for viability as described in Materials and Methods (Treg, CD3+CD4+CD25+FOXP3+; Ml macrophages, CD11Bhigh F4/80int).

Discussion

A main goal and challenge of radioimmunotherapy is to engender systemic immunity against distant nonirradiated metastatic lesions, known as the abscopal effect. Cotreatment with immune-potentiating drugs is essential to augment radiation-induced systemic immune responses to effectively control the growth of distant lesions. Indeed, preclinical studies in mice have demonstrated the benefits of combining radiation with immune modulation (35), although the abscopal effects were generally modest (68,10,11).

Immune-stimulatory antibodies, blocking CTLA-4 antibodies, agonistic 4–1BB antibodies, and to a lesser extent PD-1 or PD-L1 antibodies, can elicit significant toxicities inpatients (13,14), and combinatorial strategies that enhance their immune potency could further exacerbate such toxicities (15,16). In murine studies, CTLA-4 (7,10,42) and 4–1BB (3941) antibodies enhanced radiation-induced tumor immunity but, to the best of our knowledge, toxicities were not evaluated. To reduce potential toxicities, our approach was to target the immune-stimulatory drugs to the irradiated tumor lesion by conjugation to a targeting ligand that binds to products upregulated by radiation in the tumor stroma.

In this study, we show that combining local radiation of 4T1 breast carcinoma tumors with systemically administered VEGF-targeted 4–1BB costimulation elicited a potent antitumor immune response capable of controlling the growth of distant nontreated subcutaneous and metastatic 4T1 tumor lesions (Figs. 3 and4;Supplementary Fig. S4). The 4T1 breast carcinoma cell line is a highly metastatic poorly immunogenic tumor that has been notoriously difficult to treat by immunologic means (45). Treatment with VEGF-4–1BB conjugate also potentiated a radiation-induced antitumor response in the autochthonous carcinongen-induced fibrosarcoma model (Supplementary Fig. S2). In patients administration of agonistic 4–1BB antibodies was associated with severe hepatic toxicities and fatalities (20). Importantly, therapeutic doses of VEGF-targeted 4–1BB aptamer conjugates did not exhibit measurable toxicities when used in combination with radiation, whereas 4–1BB antibodies elicited CD8+ T-cell hyperplasia, T-cell activation (Fig. 2AF), and nonspecific inflammation in the lung and liver and (Fig. 2G andH). Evidence of liver dysfunction was suggested by the elevated levels of AST and ALT seen in the circulation of mice receiving local tumor radiation and 4–1BB antibody, but not or to a lesser extent VEGF-4–1BB aptamer conjugates (Fig. 2I). When used in combination with radiation, nonspecific CD8+ proliferation (Fig. 2AC) and liver inflammation [Fig. 2H, “ +4–1BB (1×)”] was also seen when using 8-fold less, nontherapeutic doses of 4–1BB antibody, not seen when 4–1BB antibody was administered in the absence of irradiation (21). This supports the view that combination therapies that enhance immune potency could also exacerbate (immune) pathology and that targeting will be essential to realize the therapeutic potential of the immunomodulatory ligands.

Given the clinical importance of checkpoint blockade (46,47), we also compared the VEGF-4–1BB conjugates to PD-1 and CTLA-4 antibodies. The VEGF-4–1BB conjugate elicited a comparable abscopal effect to that of CTLA-4 antibody when used in combination with radiation and measured 19 days postirradiation (Fig. 3B). Mice treated with CTLA-4, alone or in combination with radiation and/or PD-1 antibody exhibited significant inflammation and tissue destruction in the liver, lung (Supplementary Figs. S6 andS7), and notably in the small intestine (Fig. 5,S5), consistent with enterocolitis being the main toxicity seen in patients treated with CTLA-4 antibody (43). Strikingly, no inflammation or tissue damage was seen in the small intestine (Fig 5;Supplementary Fig. S5), nor that of liver or lung (Supplementary Figs. S6 andS7) of mice treated by radiation + VEGF-4–1BB aptamer conjugates. Local tumor irradiation combined with CTLA-4 antibodies, and especially with both CTLA-4 and PD-1 antibodies, but not with VEGF-4–1BB aptamer conjugates, led to increased levels of MCP-1, IL6, and GM-CSF in the circulation (Supplementary Fig. S8). Overall, these studies suggest that potentiating radiation-induced antitumor immunity by VEGF-targeted 4–1BB costimulation with aptamer conjugates may be superior to that of 4–1 BB antibody or CTLA-4 antibody therapy especially in terms of reduced toxicity.

The underlying rationale of RadVax has been that local tumor irradiation can stimulate immune responses through multiple mechanisms by promoting “immunogenic (tumor) cell death” (1,2), attenuate immune-suppressive mechanisms (48), or broaden the repertoire of tumor antigens recognized by the TILs (42,49). Here we exploit another feature of radiation, upregulation of VEGF, to expand the scope of tumor-targeted immune modulation to otherwise low VEGF-expressing tumors. We show that VEGF-targeted 4–1BB costimulation can be extended to VEGFlow tumors by radiation (Fig. 1C andD). Thus, local tumor radiation can serve not only as a means of generating a potent antitumor response but also as way to broaden the scope and enhance the efficiency of tumor-targeted immune modulation.

VEGF and 4–1BB are prototype stromal targets and immunomodulatory receptors, respectively, and were used in this study to demonstrate the feasibility and potential benefits of tumor stroma-targeted immune modulation in conjunction with radiation. Multiple products are upregulated in the tumor stroma, metallo-proteases, osteopontin, fibrin, many of which, like VEGF are further upregulated following irradiation (50). Likewise, other inhibitory (PD-1, Tim-3, LAG-3, TIGIT, BTLA) or stimulatory (OX40, LIGHT, CD27) receptors can be targeted in a similar fashion. It is conceivable that other forms of cytotoxic therapy like chemotherapeutic agents or molecularly targeted drugs are also likely to elevate VEGF or other products and maybe exploited for improving drug targeting. Future studies will determine the optimal choices of stromal targets, (combination of) immune ligands, and/or cytotoxic therapies.

Supplementary Material

supplementary figure

Acknowledgments

We thank Anugraha Rajagopalan and Yvonne Puplampu-Dove for help in data collection and Tal Gefen and Alexey Berezhnoy for technical assistance.

Grant Support

This work was supported by grant BC130871 from Congressionally Directed Medical Research Programs to E. Gilboa.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Footnotes

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

References

  • 1.Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer G. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell2015;28:690–714. [DOI] [PubMed] [Google Scholar]
  • 2.Golden EB, Apetoh L. Radiotherapy and immunogenic cell death. Semin Radiat Oncol2015;25:11–7. [DOI] [PubMed] [Google Scholar]
  • 3.Formenti SC, Demaria S. Combining radiotherapy and cancer immunotherapy: a paradigm shift. I Natl Cancer Inst2013;105:256–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Tang C, Wang X, Soh H, Seyedin S, Cortez MA, Krishnan S, et al. Combining radiation and immunotherapy: a new systemic therapy for solid tumors?Cancer Immunol Res2014;2:831–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Demaria S, Formenti SC. Radiation as an immunological adjuvant: current evidence on dose and fractionation. Front Oncol2012;2:153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Demaria S, Ng B, Devitt ML, Babb JS, Kawashima N, Liebes L, et al. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys2004;58:862–70. [DOI] [PubMed] [Google Scholar]
  • 7.Demaria S, Kawashima N, Yang AM, Devitt ML, Babb JS, Allison JP, et al. Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res2005:11:728–34. [PubMed] [Google Scholar]
  • 8.Park SS, Dong H, Liu X, Harrington SM, Krco CJ, Grams MP, et al. PD-1 restrains radiotherapy-induced abscopal effect. Cancer Immunol Res2015;3:610–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Vanpouille-Box C, Diamond JM, Pilones KA, Zavadil J, Babb JS, Formenti SC, et al. TGFbeta is a master regulator of radiation therapy-induced antitumor immunity. Cancer Res2015;75:2232–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Dewan MZ, Galloway AE, Kawashima N, Dewyngaert JK, Babb JS, Formenti SC, et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res2009;15:5379–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Shiraishi K, Ishiwata Y, Nakagawa K, Yokochi S, Taruki C, Akuta T, et al. Enhancement of antitumor radiation efficacy and consistent induction of the abscopal effect in mice by ECI301, an active variant of macrophage inflammatory protein-1 alpha. Clin Cancer Res2008;14:1159–66. [DOI] [PubMed] [Google Scholar]
  • 12.Crittenden M, Kohrt H, Levy R, Jones J, Camphausen K, Dicker A, et al. Current clinical trials testing combinations of immunotherapy and radiation. Semin Radiat Oncol2015;25:54–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J Clin Oncol2015;33:1974–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gangadhar TC, Vonderheide RH. Mitigating the toxic effects of anticancer immunotherapy. Nat Rev Clin Oncol2015;11:91–9. [DOI] [PubMed] [Google Scholar]
  • 15.Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med2013;369:122–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ribas A, Hodi FS, Callahan M, Konto C, Wolchok J. Hepatotoxicity with combination of vemurafenib and ipilimumab. N Engl J Med2013;368: 1365–6. [DOI] [PubMed] [Google Scholar]
  • 17.Wang C, Lin GH, McPherson AJ, Watts TH. Immune regulation by 4–1BB and 4–1BBL: complexities and challenges. Immunol Rev2009;229: 192–215. [DOI] [PubMed] [Google Scholar]
  • 18.Dubrot J, Milheiro F, Alfaro C, Palazon A, Martinez-Forero I, Perez-Gracia JL, et al. Treatment with anti-CD 137 mAbs causes intense accumulations of liver T cells without selective antitumor immunotherapeutic effects in this organ. Cancer Immunol Immunother2010;59:1223–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Niu L, Strahotin S, Hewes B, Zhang B, Zhang Y, Archer D, et al. Cytokine-mediated disruption of lymphocyte trafficking, hemopoiesis, and induction of lymphopenia, anemia, and thrombocytopenia in anti-CD 137-treated mice. J Immunol2007;178:4194–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Yonezawa A, Dutt S, Chester C, Kim J, Kohrt HE. Boosting cancer immunotherapy with anti-CD137 antibody therapy. Clin Cancer Res2015; 21:3113–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Schrand B, Berezhnoy A, Brenneman R, Williams A, Levay A, Kong LY, et al. Targeting 4–1BB costimulation to the tumor stroma with bispecific aptamer conjugates enhances the therapeutic index of tumor immunotherapy. Cancer Immunol Res2014;2:867–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhou J, Rossi JJ. Cell-type-specific, Aptamer-functionalized agents for targeted disease therapy. Mol Ther Nucleic Acids2014;3:e169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ferrara NVascular endothelial growth factor: basic science and clinical progress. Endocr Rev2004;25:581–611. [DOI] [PubMed] [Google Scholar]
  • 24.Senger DR, Van de Water L, Brown LF, Nagy JA, Yeo KT, Yeo TK, et al. Vascular permeability factor (VPF, VEGF) in tumor biology. Cancer Metastasis Rev1993;12:303–24. [DOI] [PubMed] [Google Scholar]
  • 25.Borre M, Nerstrom B, Overgaard J. Association between immunohistochemical expression of vascular endothelial growth factor (VEGF), VEGF-expressing neuroendocrine-differentiated tumor cells, and outcome in prostate cancer patients subjected to watchful waiting. Clin Cancer Res2000;6:1882–90. [PubMed] [Google Scholar]
  • 26.Oosting SF, Brouwers AH, van Es SC, Nagengast WB, Oude Munnink TH, Lub-de Hooge MN, et al. 89Zr-bevacizumab PET visualizes heterogeneous tracer accumulation in tumor lesions of renal cell carcinoma patients and differential effects of antiangiogenic treatment. J Nucl Med2015;56:63–69. [DOI] [PubMed] [Google Scholar]
  • 27.Yudoh K, Kanamori M, Ohmori K, Yasuda T, Aoki M, Kimura T. Concentration of vascular endothelial growth factor in the tumour tissue as a prognostic factor of soft tissue sarcomas. Br J Cancer2001;84:1610–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Shih CH, Ozawa S, Ando N, Ueda M, Kitajima M. Vascular endothelial growth factor expression predicts outcome and lymph node metastasis in squamous cell carcinoma of the esophagus. Clin Cancer Res2000; 6:1161–8. [PubMed] [Google Scholar]
  • 29.Zlobec I, Steele R, Compton CC. VEGF as a predictive marker of rectal tumor response to preoperative radiotherapy. Cancer2005;104:2517–21. [DOI] [PubMed] [Google Scholar]
  • 30.Barker HE, Paget JT, Khan AA, Harrington IQ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer2015;15:409–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wang J, Boerma M, Fu Q, Hauer-lensen M. Significance of endothelial dysfunction in the pathogenesis of early and delayed radiation enteropathy. World J Gastroenterol2007;13:3047–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kaliski A, Maggiorella L, Cengel KA, Mathe D, Rouffiac V, Opolon P, et al. Angiogenesis and tumor growth inhibition by a matrix metalloproteinase inhibitor targeting radiation-induced invasion. Mol Cancer Ther2005; 4:1717–28. [DOI] [PubMed] [Google Scholar]
  • 33.Gorski DH, Beckett MA, laskowiak NT, Calvin DP, Mauceri HJ, Salloum RM, et al. Blockage of the vascular endothelial growth factor stress response increases the antitumor effects of ionizing radiation. Cancer Res1999; 59:3374–78. [PubMed] [Google Scholar]
  • 34.Solberg TD, Nearman J, Mullins J, Li S, Baranowska-Kortylewicz J. Correlation between tumor growth delay and expression of cancer and host VEGF, VEGFR2, and osteopontin in response to radiotherapy. Int J Radiat Oncol Biol Phys2008;72:918–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Nozue M, Isaka N, Fukao K. Over-expression of vascular endothelial growth factor after preoperative radiation therapy for rectal cancer. Oncol Rep2001;8:1247–9. [DOI] [PubMed] [Google Scholar]
  • 36.Shintani S, Kiyota A, Mihara M, Nakahara Y, Terakado N, Ueyama Y, et al. Association of preoperative radiation effect with tumor angiogenesis and vascular endothelial growth factor in oral squamous cell carcinoma. Jpn J Cancer Res2000;91:1051–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Karamanolis G, Delladetsima I, Kouloulias V, Papaxoinis K, Panayiotides I, Haldeopoulos D, et al. Increased expression of VEGF and CD31 in postradiation rectal tissue: implications for radiation proctitis. Mediators Inflamm2013;2013:515048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Burmeister PE, Lewis SD, Silva RF, Preiss JR, Horwitz LR, Pendergrast PS, et al. Directin vitro selection of a 2’-O-methyl aptamer to VEGF. Chem Biol2005;12:25–33. [DOI] [PubMed] [Google Scholar]
  • 39.Belcaid Z, Phallen JA, Zeng J, See AP, Mathios D, Gottschalk C, et al. Focal radiation therapy combined with 4–1BB activation and CTLA-4 blockade yields long-term survival and a protective antigen-specific memory response in a murine glioma model. PLoS One2014;9:el01764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Verbrugge I, Hagekyriakou J, Sharp LL, Galli M, West A, McLaughlin NM, et al. Radiotherapy increases the permissiveness of established mammary tumors to rejection by immunomodulatory antibodies. Cancer Res2012; 72:3163–74. [DOI] [PubMed] [Google Scholar]
  • 41.Newcomb EW, Lukyanov Y, Kawashima N, Alonso-Basanta M, Wang SC, Liu M, et al. Radiotherapy enhances antitumor effect of anti-CD137 therapy in a mouse Glioma model. Radiat Res2010;173:426–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature2015;520: 373–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Beck KE, Blansfield JA, Tran KQ, Feldman AL, Hughes MS, Royal RE, et al. Enterocolitis in patients with cancer after antibody blockade of cytotoxic T-lymphocyte-associated antigen 4. J Clin Oncol2006;24: 2283–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Nishikawa H, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Curr Opin Immunol2014;27:1–7. [DOI] [PubMed] [Google Scholar]
  • 45.Heppner GH, Miller FR, Shekhar PM. Nontransgenic models of breast cancer. Breast Cancer Res2000;2:331–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell2015; 161: 205–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Topalian SL, Drake CG, Pardoll DMImmune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell2015; 27:450–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama N, et al. Low-dose irradiation programs macrophage differentiation to an iNOS(+)/Ml phenotype that orchestrates effective T cell immunotherapy. Cancer Cell2013;24:589–602. [DOI] [PubMed] [Google Scholar]
  • 49.Reits EA, Hodge JW, Herberts CA, Groothuis TA, Chakraborty M, Wansley EK, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med2006;203:1259–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hallahan DE. Radiation-mediated gene expression in the pathogenesis of the clinical radiation response. Semin Radiat Oncol2006;6: 250–67. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

supplementary figure

ACTIONS

RESOURCES


[8]ページ先頭

©2009-2025 Movatter.jp