GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Association for Cancer Research (AACR)  (8)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Immunology Research Vol. 8, No. 4_Supplement ( 2020-04-01), p. A08-A08
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 8, No. 4_Supplement ( 2020-04-01), p. A08-A08
    Abstract: Background: Treatment of multiple tumors and metastasis remains a challenge for current therapeutic approaches in cancer treatment. Some responses in noninjected metastases were described in the middle of the last century after using local radiotherapy in breast cancer patients. This phenomenon was named abscopal effect. Nowadays, this effect is well known to be elicited by systemic adaptive immune responses against tumor cells. Of note, it has not been fully reproducible in the clinic mainly due to the suppressive microenvironment surrounding cancer cells. The systemic use of immune checkpoint inhibitors, such as anti-CTLA-4 and anti-PD-1, has shown promise in the treatment of many tumors, but a minority of patients respond. Oncolytic virotherapy is a promising strategy that has emerged in the last few decades. It is able to prime the host immune system against tumor epitopes, thus theoretically complementing aPD1 therapy in an appealing manner. The challenge with oncolytic immunotherapy is that not all metastases can usually be injected. Therefore, it is of importance to study if local treatment can induce distant responses. While our previous work has shown synergy between oncolytic adenovirus and aPD1, here we sought to establish if the synergy extends to noninjected tumors. Aim: In the present study we aimed to assess if local adenovirus injection can impact also noninjected tumors in animals receiving aPD1 therapy. Methods: We utilized a murine melanoma model (B16.OVA) in which each mouse has two tumors, where only one tumor receives local virotherapy and the secondary tumor is studied for possible systemic effects following local injection. Results: Improvement in the overall survival was seen in the group using both therapies (aPD-1 plus virus) compared to the single therapies alone. As well, secondary tumor growth was better controlled in that group. Further information, including the final survival curve, tumor growth and immunologic mechanism-of-action data, will be presented. Conclusion: These two cancer immunotherapeutics seem to be a promising dynamic approach that may increase the life expectancy and cure the patients suffering from metastatic cancer when translated to the clinic, besides minimizing considerably the toxicity due to the specificity of such approach, and finally improving patients’ quality of life during the treatment. Citation Format: Dafne C.A. Quixabeira, Víctor Cervera-Carrascón, Riikka Havunen, Mikko Siurala, Akseli Hemminki. Adenovirus coding for TNF-α and IL-2 proteins mediates abscopal effect in mice receiving anti-PD1 immunotherapy [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27-30; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(4 Suppl):Abstract nr A08.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Immunology Research Vol. 3, No. 10_Supplement ( 2015-10-01), p. B18-B18
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 3, No. 10_Supplement ( 2015-10-01), p. B18-B18
    Abstract: Adoptive cell transfer (ACT) has shown promising results in clinical trials for melanoma, with response rates ranging from 40% to 72%. We aimed to enhance the efficacy of ACT by coupling it with adenoviruses coding for immunostimulatory cytokines. The non-replicating adenoviruses that were constructed (Ad5-CMV-mIFNg, Ad5-CMV-mIL2, Ad5-CMV-mIFNb and Ad5-CMV-mTNFa) were confirmed to produce biologically active murine cytokines in vitro. These viruses were then used in combination with adoptive transfer of OT-1 TCR transgenic T-cells to treat C57BL/6 mice bearing B16-OVA melanoma tumors. The animals were administered intraperitoneally with 1.5 x 106 CD8+-enriched OT-1 cells with or without intratumoral injections of cytokine-coding adenoviruses (1 x 109 viral particles/tumor). Combination treatment with Ad5-CMV-mIL2 and OT-1 resulted in statistically significant antitumor efficacy when compared with either monotherapy and untreated control (combination vs. virus, p=0.026; combination vs. OT1, p=0.003; combination vs. mock, p & lt;0.001). Similar results were obtained with other cytokine-coding virus and OT-1 combinations, except for mIFNb. In further experiments a triple combination of Ad5-CMV-mIL2 + Ad5-CMV-mTNFa (1:1 ratio) and OT-1 T-cells were used to treat B16-OVA; improved antitumor efficacy was observed with the triple combination over dual agent therapies (triple combination vs. Ad5-CMV-mIL2 + OT-1, p=0.001; triple combination vs. Ad5-CMV-mTNFa + OT-1, p=0.049). In summary, these results support the further development of a novel immunotherapeutic approach for the treatment of melanoma. Citation Format: Mikko Siurala, Riikka Havunen, Simona Bramante, Dipongkor Saha, Siri Tähtinen, Markus Vähä-Koskela, Akseli Hemminki. Adoptive T-cell transfer combined with murine cytokine-armed adenoviruses for the treatment of melanoma. [abstract]. In: Proceedings of the AACR Special Conference: Tumor Immunology and Immunotherapy: A New Chapter; December 1-4, 2014; Orlando, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2015;3(10 Suppl):Abstract nr B18.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Immunology Research Vol. 3, No. 10_Supplement ( 2015-10-01), p. B07-B07
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 3, No. 10_Supplement ( 2015-10-01), p. B07-B07
    Abstract: Adenovirus vectors are the most commonly used gene therapy vectors in clinical trials. Administrated systemically or intratumorally the virus is capable of inducing immune responses against the tumor per se, but this effect can also be amplified by arming the viruses with immune system stimulating factors. Moreover, we have discovered that adenoviruses enhance adoptive T-cell therapy in a potent manner. Our previous data also shows that the most promising factors to stimulate immune system are interleukin (IL) -2 and Tumor Necrosis Factor alpha (TNFα). IL-2 is a commonly used cytokine in treating malignant melanoma and renal cell carcinoma. However, systemic IL-2 can in some cases lead to severe side effects. Like IL-2, TNFα activates immune cells, but it is also known for its anti-tumor properties and it is for example capable of promoting tumor cell death by apoptosis and necrosis. Arming oncolytic viruses with cytokines results in long lasting, high level expression locally but low level systemically. Currently, we are developing oncolytic adenoviruses with expression of one or both of the above mentioned human cytokines. The viruses have an Ad5/3 chimera that is generated through replacement of the Ad5 fiber knob with the Ad3 knob while the Ad5 shaft and tail are retained. The chimera has proven to have an improved cancer cell transduction as well as antitumor efficacy. In addition, the viral genome has an E2F promoter and a 24-basepair deletion at the E1A domain, which makes it selective to retinoblastoma protein defective cells (most tumor cells). The viral particles are produced in A549 lung adenocarcinoma cell line. The genomes are sequenced and the functionality of the viruses and the cytokines proven with various methods. The secretion of cytokines is tested with ELISA and by using indicator cell lines. The functionality of the viruses is studied by infecting different cells lines with variable amount of viral particles and measuring the proportion of surviving cells with MTS-assay. Our data proves oncolytic and cytokine production capability of Ad5/3-E2F-D24-hTNFα/hIL2 adenoviruses. The results indicate that these viruses can be used in further investigations in vivo. Citation Format: Riikka Havunen, Suvi Parviainen, Mikko Siurala, Akseli Hemminki. Characterization of T-cell therapy enhancing oncolytic adenoviruses with human cytokine expression. [abstract]. In: Proceedings of the AACR Special Conference: Tumor Immunology and Immunotherapy: A New Chapter; December 1-4, 2014; Orlando, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2015;3(10 Suppl):Abstract nr B07.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 4, No. 11_Supplement ( 2016-11-01), p. B063-B063
    Abstract: Adoptive T-cell based therapy can promote dramatic tumor regressions in both pre-clinical tumor models and in patients with CD19+ hematological tumors. However, in the case of human solid tumors, anti-tumor effects are limited by T-cell tolerance and the immunosuppressive tumor microenvironment. Human data from cancer patients treated with oncolytic adenovirus indicated that lymphocytes traffic to tissues following virotherapy. Therefore, we hypothesized to enable successful T-cell therapy by arming viruses with immunostimulatory cytokines which can counteract tumor immunosuppression locally. Previously, we have identified interleukin-2 (IL-2) and Tumor Necrosis Factor alpha (TNFa) as the most promising factors to stimulate the graft used in adoptive T-cell therapy. Also, we have established that our arming approach results in long lasting, high level cytokine expression locally but low levels systemically in vivo and in patients, which is important for safety versus efficacy. One attractive aspect of this approach is the ease of combinations with standard therapies including checkpoint inhibiting antibodies (ongoing experiments). Non-replicating adenovirus vectors coding for murine TNFa and IL-2 were used together with intraperitoneally administered OT-I TCR transgenic T-cells in the murine B16.OVA melanoma model. The best antitumor efficacy was obtained when virally coded IL-2 and TNFa were both used with T-cell transfer. Moreover, IL-2/TNFa virus combination treatment was able to attract (111)In-labelled OT-I cells into B16.OVA tumors in SPECT/CT imaging experiments. In addition to mouse studies, we developed oncolytic adenoviruses expressing human TNFa and/or human IL-2 that were used in combination with intratumoral adoptive transfer of tumor-infiltrating lymphocytes (TIL) in immunocompetent and adenovirus-permissive Syrian hamsters. These viruses improved the efficacy of TIL therapy and protected animals from tumor rechallenge. Furthermore, we have shown that adenovirally delivered IL-2 is superior to systemically administered IL-2 with regard to antitumor efficacy and safety, in both mice and hamsters. TILT Biotherapeutics is in the process of confirming the results in clinical trials. Citation Format: Mikko Siurala, Riikka Havunen, João Santos, Siri Tähtinen, Dipongkor Saha, Markus Vähä-Koskela, Michael Behr, Dirk M. Nettelbeck, Anja Ehrhardt, Suvi Parviainen, Akseli Hemminki. Oncolytic adenoviruses armed with tumor necrosis factor alpha and interleukin-2 enable successful adoptive T cell therapy of solid tumors [abstract]. In: Proceedings of the Second CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; 2016 Sept 25-28; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(11 Suppl):Abstract nr B063.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 629-629
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 629-629
    Abstract: Background. Breast cancer is the most common cancer in women worldwide. Bone morphogenetic proteins (BMPs), members of the transforming growth factor β superfamily, are known to regulate cell proliferation, differentiation and motility, and have also been shown to be involved in cancer pathogenesis, also in breast cancer. We have previously demonstrated that BMP4 is able to consistently reduce breast cancer cell proliferation through G1 cell cycle arrest and to simultaneously induce migration and invasion in a subset of breast cancer cell lines. Similarly, our clinical data revealed a correlation between elevated BMP4 expression in primary breast tumors and reduced proliferation as well as increased risk of recurrence. The growth inhibitory effects of BMP4 have also been demonstrated in vivo but its possible metastasis promoting functions are less well characterized. Here we set out to investigate this topic using a xenograft mouse model. Methods. MDA-MB-231 breast cancer cells were transduced with a luciferase-expressing vector to allow monitoring of the metastasis formation using bioluminescence imaging. Cells (2 × 105) were injected into the mice intracardially and BMP4 (100 ng/g, 10 animals) or vehicle control (11 animals) was administered through tail vein three times a week. After seven weeks, the mice were sacrificed and metastases collected for histological analyses. Results. The overall amount of metastases was similar in both groups (13 in BMP4-treatment group vs. 12 in control group). There was a slight but non-significant trend of metastases developing earlier in the BMP4 group compared to controls. Most of the metastases occurred in bone and adrenal glands. There were somewhat more metastases in bone in the BMP4-treated mice (10 vs. 7) and more adrenal gland metastases in vehicle-treated animals (5 vs. 1). To assess the contribution of BMP4 to the characteristics of the metastases, the tumors were stained for pSMAD1/5/9 (BMP signaling activation), Ki67 (proliferation), MECA32 (blood vessels), mesenchymal marker vimentin, α-SMA (cancer-associated fibroblasts) and basal markers K5 and K14. No major dissimilarities were observed between the BMP4 and vehicle tumor groups in the staining patterns. Interestingly, the osteoclast marker Tartrate-resistant acid phosphatase (TRAP) was expressed in both groups in the cancer cells whereas Toluidine Blue staining revealed that the bone morphology was not detrimentally affected by BMP4 treatment. Conclusions. Despite its ability to enhance breast cancer cell migration and invasion in vitro, BMP4 does not seem to have a dramatic impact on in vivo metastasis formation, although a small acceleration in appearance of the metastases was observed. However, the limitations of the xenograft model do not allow us to exclude the possible long-term effects of BMP4 that might be more applicable to human situation. Citation Format: Minna Ampuja, Emma L. Alarmo, Philip Owens, Riikka Havunen, Agnes E. Gorska, Harold L. Moses, Anne Kallioniemi. The impact of BMP4 on breast cancer metastasis in an in vivo xenograft mouse model. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 629.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Immunology Research Vol. 8, No. 4_Supplement ( 2020-04-01), p. A30-A30
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 8, No. 4_Supplement ( 2020-04-01), p. A30-A30
    Abstract: Adenoviral-mediated gene therapy is a promising new treatment for malignant tumors with poor responses to traditional cancer treatments. In this study, we investigate the effects of danger- and pathogen-associated molecular pattern (DAMP and PAMP) signaling responses to a chimeric, cytokine armed, oncolytic adenovirus (Ad5/3-E2F-d24-hTNFα-IRES-hIL2). It has previously been shown that this virus has a two-part mode of action against tumors. It simultaneously acts as an immunostimulatory agent while the virus itself lyses cancer cells. However, as this reaction is not fully understood on a molecular level, more in-depth information on the adenovirus-induced signaling cascades in the tumor microenvironment is needed. There are several pattern recognition receptors (PRR) in cells that are responsible for inducing the signaling cascades and consequent immune response towards DAMPs and PAMPs present during adenovirus infections. It has previously been reported that the most important PRRs and sensors include TLR9, NLRP3, RIG-1, cGAS, and AIM2. Binding of DAMPs and PAMPs to PRRs results in the activation of signaling cascades consisting of a plethora of proteins, finally culminating in the production and release of cytokines, pyroptosis or cellular activation and extracellular signaling, consequently affecting neighboring cells as well. In this study, several methods such as Western blot, flow cytometry and RNA-sequencing were used to study DAMP and PAMP signaling and the subsequent reactions in the tumor microenvironment. The knowledge gained about the signaling pathways in this study provides new insight into cell signaling, which can be used to further develop immuno- and virotherapy and to provide a new method of treating poorly responsive tumors. Citation Format: Camilla Heiniö, Riikka Havunen, Mikko Siurala, Akseli Hemminki. Molecular insight into pathogen-associated molecular pattern signaling during TNFa and IL2 armed oncolytic adenovirus treatments [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27-30; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(4 Suppl):Abstract nr A30.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 8, No. 4_Supplement ( 2020-04-01), p. B07-B07
    Abstract: Dendritic cell (DC) therapy is considered a promising immunotherapeutic approach for treatment of advanced cancer. However, the immunosuppressive tumor microenvironment leads to DC dysfunction. Therefore, in clinical trials DC therapy has generally failed to fulfill its expectations. Oncolytic adenoviruses are well tolerated and have shown to activate antitumor immune responses. Importantly, they can convert immunosuppression locally into a proinflammatory state. To improve the proper activation of transferred DCs, we armed oncolytic adenovirus with CD40 ligand (CD40L). CD40L is known to play an important role in the regulation of immune cells through its capacity to stimulate dendritic cells that leads to the activation of cytotoxic T cells. Therefore, we generated a novel virus Ad3-hTERT-CMV-hCD40L, which is fully serotype 3 adenovirus (Ad3) for intravenous delivery. It features a human telomerase reverse transcriptase (hTERT) promoter for tumor specific replication and expresses human CD40L (hCD40L) under a cytomegalovirus (CMV) promoter for induction of antitumor efficacy. Of note, human and animal data have shown the ability of Ad3 to successfully reach tumors through the intravenous route. In syngeneic studies in an immunocompetent model, DC therapy with our murine CD40L-armed adenovirus showed significant antitumor immune response. This enhanced therapeutic effect is associated with increased tumor specific T cells and induction of T-helper type 1 immune response. This synergistic effect was further evaluated in mice humanized with human peripheral blood mononuclear cells. Treatment with hCD40L-armed adenovirus and human DCs showed 100% survival in conjunction with tumor control. To conclude, CD40L armed oncolytic adenovirus 3 improves DC therapy by favorable alteration of tumor microenvironment. These findings support clinical trials where DC therapy is enhanced with oncolytic adenovirus. Citation Format: João Manuel Santos, Akseli Hemminki, Mikko Siurala, Otto Hemminki, Riikka Havunen, Victor Cervera-Carrascon, Suvi Sorsa, Hongjie Wang, Andre Lieber, Tanja de Gruijl, Anna Kanerva, Sadia Zafar. Oncolytic adenovirus 3 coding for CD40L as an enhancer of dendritic cell therapy [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27-30; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(4 Suppl):Abstract nr B07.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Immunology Research Vol. 8, No. 4_Supplement ( 2020-04-01), p. B16-B16
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 8, No. 4_Supplement ( 2020-04-01), p. B16-B16
    Abstract: Checkpoint inhibitors (CPI) started a new golden era of immunotherapy for cancer treatment. Since approval of the first product in 2011, a number of these antibodies are now available for different indications. Hundreds of trials are ongoing, which could predict many further approvals soon. Unfortunately, only a minority of patients currently benefit from CPI treatment. One of the biggest limitations appears to be poor immune infiltration of many tumors, known as immune desert or excluded tumors. Oncolytic viruses are an interesting tool to use together with CPI to increase the frequency and the quality of patients’ responses. Besides their inherent ability to raise the immune alarm towards the tumor, resulting in recruitment of lymphocytes and other cells, they can be armed with transgenes to potentiate these effects. In a data-driven approach, adenoviruses expressing Tumor Necrosis Factor Alpha and Interleukin 2 were identified as the optimal approach for recruiting and activating T cells in tumors. In this approach, the regimen of administration must be considered as it can affect the outcome of the treatment. We used an aggressive and immunosuppressive solid tumor model (B16.OVA melanoma) to study the interaction between the above-mentioned viruses and anti-PD-1 and anti-PD-L1. After optimizing treatment regimes, control of every tumor led to complete responses in 100% of the animals when the viral platform and anti-PD-1 (or anti-PD-L1) were used together. This was a significant improvement when compared to them used as monotherapies (p-value & lt;0.01), but the optimal regimens of administration were different for anti-PD-1 and for anti-PD-L1. Full data, including tumor growth control, survival and extensive immunologic characterization, revealing the mechanism of action, will be presented. Checkpoint inhibitors and oncolytic adenovirus are an attractive approach for increasing the proportion of patients benefiting from CPI therapy. It is of critical relevance to understand how to best use them to maximize their therapeutic effect. Our study underlines the importance of scheduling of the agents with each other, and the differences between CPI targets. Citation Format: Victor Cervera-Carrascon, Dafne Quixabeira, Eleonora Munaro, Joao M. Santos, Riikka Havunen, Mikko Siurala, Akseli Hemminki. Enabling checkpoint inhibitors with oncolytic viruses to deliver complete responses: A matter of timing [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27-30; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(4 Suppl):Abstract nr B16.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...