Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Riikka Havunen is active.

Publication


Featured researches published by Riikka Havunen.


OncoImmunology | 2017

Intravenously usable fully serotype 3 oncolytic adenovirus coding for CD40L as an enabler of dendritic cell therapy

Sadia Zafar; Suvi Parviainen; Mikko Siurala; Otto Hemminki; Riikka Havunen; Siri Tähtinen; Simona Bramante; Lotta Vassilev; Hongjie Wang; André Lieber; Silvio Hemmi; Tanja D. de Gruijl; Anna Kanerva; Akseli Hemminki

ABSTRACT Vaccination with dendritic cells (DCs), the most potent professional antigen-presenting cells in the body, is a promising approach in cancer immunotherapy. However, tumors induce immunosuppression in their microenvironment that suppresses and impairs the function of DCs. Therefore, human clinical trials with DC therapy have often been disappointing. To improve the therapeutic efficacy and to overcome the major obstacles of DC therapy, we generated a novel adenovirus, Ad3-hTERT-CMV-hCD40L, which is fully serotype 3 and expresses hCD40L for induction of antitumor immune response. The specific aim is to enhance DCs function. Data from a human cancer patient indicated that this capsid allows effective transduction of distant tumors through the intravenous route. Moreover, patient data suggested that virally produced hCD40L can activate DCs in situ. The virus was efficient in vitro and had potent antitumor activity in vivo. In a syngeneic model, tumors treated with Ad5/3-CMV-mCD40L virus plus DCs elicited greater antitumor effect as compared with either treatment alone. Moreover, virally coded CD40L induced activation of DCs, which in turn, lead to the induction of a Th1 immune response and increased tumor-specific T cells. In conclusion, Ad3-hTERT-CMV-hCD40L is promising for translation into human trials. In particular, this virus could enable successful dendritic cell therapy in cancer patients.


Molecular Therapy - Oncolytics | 2017

Oncolytic Adenoviruses Armed with Tumor Necrosis Factor Alpha and Interleukin-2 Enable Successful Adoptive Cell Therapy

Riikka Havunen; Mikko Siurala; Suvi Sorsa; Susanna Grönberg-Vähä-Koskela; Michael Behr; Siri Tähtinen; João Manuel Santos; Pauliina Karell; Juuso Rusanen; Dirk M. Nettelbeck; Anja Ehrhardt; Anna Kanerva; Akseli Hemminki

Adoptive cell therapy holds much promise in the treatment of cancer but results in solid tumors have been modest. The notable exception is tumor-infiltrating lymphocyte (TIL) therapy of melanoma, but this approach only works with high-dose preconditioning chemotherapy and systemic interleukin (IL)-2 postconditioning, both of which are associated with toxicities. To improve and broaden the applicability of adoptive cell transfer, we constructed oncolytic adenoviruses coding for human IL-2 (hIL2), tumor necrosis factor alpha (TNF-α), or both. The viruses showed potent antitumor efficacy against human tumors in immunocompromised severe combined immunodeficiency (SCID) mice. In immunocompetent Syrian hamsters, we combined the viruses with TIL transfer and were able to cure 100% of the animals. Cured animals were protected against tumor re-challenge, indicating a memory response. Arming with IL-2 and TNF-α increased the frequency of both CD4+ and CD8+ TILs in vivo and augmented splenocyte proliferation ex vivo, suggesting that the cytokines were important for T cell persistence and proliferation. Cytokine expression was limited to tumors and treatment-related signs of systemic toxicity were absent, suggesting safety. To conclude, cytokine-armed oncolytic adenoviruses enhanced adoptive cell therapy by favorable alteration of the tumor microenvironment. A clinical trial is in progress to study the utility of Ad5/3-E2F-d24-hTNFa-IRES-hIL2 (TILT-123) in human patients with cancer.


Molecular Therapy | 2016

Adenoviral Delivery of Tumor Necrosis Factor-α and Interleukin-2 Enables Successful Adoptive Cell Therapy of Immunosuppressive Melanoma

Mikko Siurala; Riikka Havunen; Dipongkor Saha; Dave Lumen; Anu J. Airaksinen; Siri Tähtinen; Víctor Cervera-Carrascon; Simona Bramante; Suvi Parviainen; Markus Vähä-Koskela; Anna Kanerva; Akseli Hemminki

Adoptive T-cell transfer is a promising treatment approach for metastatic cancer, but efficacy in solid tumors has only been achieved with toxic pre- and postconditioning regimens. Thus, adoptive T-cell therapies would benefit from complementary modalities that enable their full potential without excessive toxicity. We aimed to improve the efficacy and safety of adoptive T-cell transfer by using adenoviral vectors for direct delivery of immunomodulatory murine cytokines into B16.OVA melanoma tumors with concomitant T-cell receptor transgenic OT-I T-cell transfer. Armed adenoviruses expressed high local and low systemic levels of cytokine when injected into B16.OVA tumors, suggesting safety of virus-mediated cytokine delivery. Antitumor efficacy was significantly enhanced with adenoviruses coding for murine interleukin-2 (mIL-2) and tumor necrosis factor-α (mTNFα) when compared with T-cell transfer alone or viruses alone. Further improvement in efficacy was achieved with a triple combination of mIL-2, mTNFα, and OT-I T-cells. Mechanistic studies suggest that mIL-2 has an important role in activating T-cells at the tumor, while mTNFα induces chemokine expression. Furthermore, adenovirus treatments enhanced tumor-infiltration of OT-I T-cells as demonstrated by SPECT/CT imaging of (111)In-labeled cells. Our results suggest the utility of cytokine-coding adenoviruses for improving the efficacy of adoptive T-cell therapies.


OncoImmunology | 2016

Syngeneic Syrian hamster tumors feature tumor-infiltrating lymphocytes allowing adoptive cell therapy enhanced by oncolytic adenovirus in a replication permissive setting

Mikko Siurala; Markus Vähä-Koskela; Riikka Havunen; Siri Tähtinen; Simona Bramante; Suvi Parviainen; J. Michael Mathis; Anna Kanerva; Akseli Hemminki

ABSTRACT Adoptive transfer of tumor-infiltrating lymphocytes (TIL) has shown promising yet sometimes suboptimal results in clinical trials for advanced cancer, underscoring the need for approaches improving efficacy and safety. Six implantable syngeneic tumor cell lines of the Syrian hamster were used to initiate TIL cultures. TIL generated from tumor fragments cultured in human interleukin-2 (IL-2) for 10 d were adoptively transferred into tumor-bearing hamsters with concomitant intratumoral injections of oncolytic adenovirus (Ad5-D24) for the assessment of antitumor efficacy. Pancreatic cancer (HapT1) and melanoma (RPMI 1846) TIL exhibited potent and tumor-specific cytotoxicity in effector-to-target (E/T) assays. MHC Class I blocking abrogated the cell killing of RPMI 1846 TIL, indicating cytotoxic CD8+ T-cell activity. When TIL were combined with Ad5-D24 in vitro, HapT1 tumor cell killing was significantly enhanced over single agents. In vivo, the intratumoral administration of HapT1 TIL and Ad5-D24 resulted in improved tumor growth control compared with either treatment alone. Additionally, splenocytes derived from animals treated with the combination of Ad5-D24 and TIL killed autologous tumor cells more efficiently than monotherapy-derived splenocytes, suggesting that systemic antitumor immunity was induced. For the first time, TIL of the Syrian hamster have been cultured, characterized and used therapeutically together with oncolytic adenovirus for enhancing the efficacy of TIL therapy. Our results support human translation of oncolytic adenovirus as an enabling technology for adoptive T-cell therapy of solid tumors.


JCI insight | 2018

Pancreatic cancer therapy with combined mesothelin-redirected chimeric antigen receptor T cells and cytokine-armed oncolytic adenoviruses

Keisuke Watanabe; Yanping Luo; Tong Da; Sonia Guedan; Marco Ruella; John Scholler; Brian Keith; Regina M. Young; Boris Engels; Suvi Sorsa; Mikko Siurala; Riikka Havunen; Siri Tähtinen; Akseli Hemminki; Carl H. June

Pancreatic ductal adenocarcinoma (PDA) is characterized by its highly immunosuppressive tumor microenvironment (TME) that limits T cell infiltration and induces T cell hypofunction. Mesothelin-redirected chimeric antigen receptor T cell (meso-CAR T cell) therapy has shown some efficacy in clinical trials but antitumor efficacy remains modest. We hypothesized that combined meso-CAR T cells with an oncolytic adenovirus expressing TNF-α and IL-2 (Ad5/3-E2F-D24-TNFa-IRES-IL2, or OAd-TNFa-IL2) would improve efficacy. OAd-TNFa-IL2 enhanced the antitumor efficacy of meso-CAR T cells in human-PDA-xenograft immunodeficient mice and efficacy was associated with robustly increased tumor-infiltrating lymphocytes (TILs), enhanced and prolonged T cell function. Mice treated with parental OAd combined with meso-CAR T developed tumor metastasis to the lungs even if primary tumors were controlled. However, no mice treated with combined OAd-TNFa-IL2 and meso-CAR T died of tumor metastasis. We also evaluated this approach in a syngeneic mouse tumor model by combining adenovirus expressing murine TNF-α and IL-2 (Ad-mTNFa-mIL2) and mouse CAR T cells. This approach induced significant tumor regression in mice engrafted with highly aggressive and immunosuppressive PDA tumors. Ad-mTNFa-mIL2 increased both CAR T cell and host T cell infiltration to the tumor and altered host tumor immune status with M1 polarization of macrophages and increased dendritic cell maturation. These findings indicate that combining cytokine-armed oncolytic adenovirus to enhance the efficacy of CAR T cell therapy is a promising approach to overcome the immunosuppressive TME for the treatment of PDA.


OncoImmunology | 2018

TNFa and IL-2 armed adenoviruses enable complete responses by anti-PD-1 checkpoint blockade

Víctor Cervera-Carrascon; Mikko Siurala; João Manuel Santos; Riikka Havunen; Siri Tähtinen; Pauliina Karell; Suvi Sorsa; Anna Kanerva; Akseli Hemminki

ABSTRACT Releasing the patients immune system against their own malignancy by the use of checkpoint inhibitors is delivering promising results. However, only a subset of patients currently benefit from them. One major limitation of these therapies relates to the inability of T cells to detect or penetrate into the tumor resulting in unresponsiveness to checkpoint inhibition. Virotherapy is an attractive tool for enabling checkpoint inhibitors as viruses are naturally recognized by innate defense elements which draws the attention of the immune system. Besides their intrinsic immune stimulating properties, the adenoviruses used here are armed to express tumor necrosis factor alpha (TNFa) and interleukin-2 (IL-2). These cytokines result in immunological danger signaling and multiple appealing T-cell effects, including trafficking, activation and propagation. When these viruses were injected into B16.OVA melanoma tumors in animals concomitantly receiving programmed cell-death protein 1 (PD-1) blocking antibodies both tumor growth control (p < 0.0001) and overall survival (p < 0.01) were improved. In this set-up, the addition of adoptive cell therapy with OT-I lymphocytes did not increase efficacy further. When virus injections were initiated before antibody treatment in a prime-boost approach, 100% of tumors regressed completely and all mice survived. Viral expression of IL2 and TNFa altered the cytokine balance in the tumor microenvironment towards Th1 and increased the intratumoral proportion of CD8+ and conventional CD4+ T cells. These preclinical studies provide the rationale and schedule for a clinical trial where oncolytic adenovirus coding for TNFa and IL-2 (TILT-123) is used in melanoma patients receiving an anti-PD-1 antibody.


International Journal of Cancer | 2017

Adenoviral production of interleukin‐2 at the tumor site removes the need for systemic postconditioning in adoptive cell therapy

João Manuel Santos; Riikka Havunen; Mikko Siurala; Víctor Cervera-Carrascon; Siri Tähtinen; Suvi Sorsa; Marjukka Anttila; Pauliina Karell; Anna Kanerva; Akseli Hemminki

Systemic high dose interleukin‐2 (IL‐2) postconditioning has long been utilized in boosting the efficacy of T cells in adoptive cell therapy (ACT) of solid tumors. The resulting severe off‐target toxicity of these regimens renders local production at the tumor an attractive concept with possible safety gains. We evaluated the efficacy and safety of intratumorally administered IL‐2‐coding adenoviruses in combination with tumor‐infiltrating lymphocyte therapy in syngeneic Syrian hamsters bearing HapT1 pancreatic tumors and with T cell receptor transgenic ACT in B16.OVA melanoma bearing C57BL/6 mice. The models are complementary: hamsters are semi‐permissive for human oncolytic adenovirus, whereas detailed immunological analyses are possible in mice. In both models, local production of IL‐2 successfully replaced the need for systemic recombinant IL‐2 (rIL‐2) administration and increased the efficacy of the cell therapy. Furthermore, vectored delivery of IL‐2 significantly enhanced the infiltration of CD8+ T cells, M1‐like macrophages, and B‐cells while systemic rIL‐2 increased CD25 + FoxP3+ T cells at the tumor. In contrast with vectored delivery, histopathological analysis of systemic rIL‐2‐treated animals revealed significant changes in lungs, livers, hearts, spleens, and kidneys. In summary, local IL‐2 production results in efficacy and safety gains in the context of ACT. These preclinical assessments provide the rationale for ongoing clinical translation.


Molecular Therapy | 2016

515. Oncolytic Adenovirus Armed with Cytokines Enhances CAR-T Cell Efficacy in Pancreatic Tumor Model

Keisuke Watanabe; John Scholler; Sonia Guedan; Shannon E. McGettigan; Yanping Luo; Siri Tähtinen; Suvi Parviainen; Riikka Havunen; Mikko Siurala; Akseli Hemminki; Carl H. June

Background: Chimeric antigen receptor T-cell (CAR-T) therapy has shown significant efficacy in hematological malignancies, however, the efficacy against solid tumors remains limited. Immunosuppression caused by tumor microenvironment or poor infiltration of transferred T cells can restrict T cell efficacy. We propose that oncolytic Adenovirus (O-Ad) armed with cytokines improves the efficacy of adoptive T cell therapy by modulating the tumor environment. Aim: Here we aimed to study if O-Ad armed with cytokines can 1) cause direct lysis of pancreatic cancer cells, 2) enhance killing by CAR-T cells. 3) enhance infiltration and persistence of CAR-T cells in the context of solid tumors. Methods: We targeted pancreatic tumor cell lines by mesothelin specific CAR-T cells (SS1-BBz CAR-T) in combination with O-Ad; Adv-5/3-d24-IL2 or Adv-5/3-d24-TNF-IL2, which consist of an adenovirus serotype 5 nucleic acid backbone, a serotype 5/3 chimeric fiber knob, a 24-bp deletion (d24) in the Rb binding constant region 2 of E1 promoter, an E2F tumor specific promoter and cytokines (interleukin 2 or tumor necrosis factor alpha or both). Results: Pancreatic tumor cell lines used in this study; ASPC1, BXPC3, Capan2 expressed the Adv-serotype 3 receptor, DSG2. We also confirmed that O-Ad does not have adverse effects on T cell viability and proliferation even at high titer (1,000vp/cell) in an in vitro assay. To look at the efficacy of the O-Ad and CAR-T combination, we performed a killing assay. O-Ad clearly enhanced killing by CAR-T cells in luciferase based assay. We also used xCELLigence real time cell analyzer (RTCA) for kinetic analysis of killing. In combination with O-Ad, more rapid killing kinetics by CAR-T cells were observed especially in lower E:T ratio. To look at the impact of the combination in vivo, subcutaneous ASPC1-CBG-GFP in NSG mice were treated with CAR-T alone or in combination with intra-tumoral injection of O-Ad. O-Ad showed significant synergy with CAR-T in luciferase based photon assay. And higher number of T cells was observed in spleen in the O-Ad armed with IL2 combination group. Conclusions: These results suggest that combination therapy of O-Ad armed with cytokine(s) and CAR-T cells is effective against solid tumors by enhancing T cell activity.


Oncotarget | 2018

Interleukin 8 activity influences the efficacy of adenoviral oncolytic immunotherapy in cancer patients

Kristian Taipale; Siri Tähtinen; Riikka Havunen; Anniina Koski; Ilkka Liikanen; Päivi Pakarinen; Riitta Koivisto-Korander; Matti Kankainen; Timo Joensuu; Anna Kanerva; Akseli Hemminki

After the landmark approval of T-VEC, oncolytic viruses are finding their way to the clinics. However, response rates have still room for improvement, and unfortunately there are currently no available markers to predict responses for oncolytic immunotherapy. Interleukin 8 (IL-8) production is upregulated in many cancers and it also connects to several pathways that have been shown to impair the efficacy of adenoviral immunotherapy. We studied the role of IL-8 in 103 cancer patients treated with oncolytic adenoviruses. We found high baseline serum IL-8 concentration to be independently associated with poor prognosis (p<0.001). Further, normal baseline IL-8 was associated with improved prognostic potential of calculation of the neutrophil-to-lymphocyte ratio (p<0.001). Interestingly, a decrease in IL-8 concentration after treatment with oncolytic adenovirus predicted better overall survival (p<0.001) and higher response rate, although this difference was not significant (p=0.066). We studied the combination of adenovirus and IL-8 neutralizing antibody ex vivo in single cell suspensions and in co-cultures of tumor-associated CD15+ neutrophils and CD3+ tumor-infiltrating lymphocytes derived from fresh patient tumor samples. These results indicate a role for IL-8 as a biomarker in oncolytic virotherapy, but additionally provide a rationale for targeting IL-8 to improve treatment efficacy. In conclusion, curtailing the activity of IL-8 systemically or locally in the tumor microenvironment could improve anti-tumor immune responses resulting in enhanced efficacy of adenoviral immunotherapy of cancer.


OncoImmunology | 2018

CD40L coding oncolytic adenovirus allows long-term survival of humanized mice receiving dendritic cell therapy

Sadia Zafar; Suvi Sorsa; Mikko Siurala; Otto Hemminki; Riikka Havunen; Víctor Cervera-Carrascon; João Manuel Santos; Hongjie Wang; André Lieber; Tanja D. de Gruijl; Anna Kanerva; Akseli Hemminki

ABSTRACT Dendritic cells (DCs) are crucial players in promoting immune responses. Logically, adoptive DC therapy is a promising approach in cancer immunotherapy. One of the major obstacles in cancer immunotherapy in general is the immunosuppressive tumor microenvironment, which hampers the maturation and activation of DCs. Therefore, human clinical outcomes with DC therapy alone have been disappointing. In this study, we use fully serotype 3 oncolytic adenovirus Ad3-hTERT-CMV-hCD40L, expressing human CD40L, to modulate the tumor microenvironment with subsequently improved function of DCs. We evaluated the synergistic effects of Ad3-hTERT-CMV-hCD40L and DCs in the presence of human peripheral blood mononuclear cells ex vivo and in vivo. Tumors treated with Ad3-hTERT-CMV-hCD40L and DCs featured greater antitumor effect compared with unarmed virus or either treatment alone. 100% of humanized mice survived to the end of the experiment, while mice in all other groups died by day 88. Moreover, adenovirally-delivered CD40L induced activation of DCs, leading to induction of Th1 immune responses. These results support clinical trials with Ad3-hTERT-CMV-hCD40L in patients receiving DC therapy.

Collaboration


Dive into the Riikka Havunen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anja Ehrhardt

Witten/Herdecke University

View shared research outputs
Top Co-Authors

Avatar

Dirk M. Nettelbeck

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge