Network


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

Hotspot


Dive into the research topics where Mari Raki is active.

Publication


Featured researches published by Mari Raki.


Molecular Therapy | 2010

Treatment of Cancer Patients With a Serotype 5/3 Chimeric Oncolytic Adenovirus Expressing GMCSF

Anniina Koski; Lotta Kangasniemi; Sophie Escutenaire; Sari Pesonen; Vincenzo Cerullo; Iulia Diaconu; Petri Nokisalmi; Mari Raki; Maria Rajecki; Kilian Guse; Tuuli Ranki; Minna Oksanen; Sirkka-Liisa Holm; Elina Haavisto; Aila Karioja-Kallio; Leena Laasonen; Kaarina Partanen; Matteo Ugolini; Andreas Helminen; Eerika Karli; Päivi Hannuksela; S Pesonen; Timo Joensuu; Anna Kanerva; Akseli Hemminki

Augmenting antitumor immunity is a promising way to enhance the potency of oncolytic adenoviral therapy. Granulocyte-macrophage colony-stimulating factor (GMCSF) can mediate antitumor effects by recruiting natural killer cells and by induction of tumor-specific CD8(+) cytotoxic T-lymphocytes. Serotype 5 adenoviruses (Ad5) are commonly used in cancer gene therapy. However, expression of the coxsackie-adenovirus receptor is variable in many advanced tumors and preclinical data have demonstrated an advantage for replacing the Ad5 knob with the Ad3 knob. Here, a 5/3 capsid chimeric and p16-Rb pathway selective oncolytic adenovirus coding for GMCSF was engineered and tested preclinically. A total of 21 patients with advanced solid tumors refractory to standard therapies were then treated intratumorally and intravenously with Ad5/3-D24-GMCSF, which was combined with low-dose metronomic cyclophosphamide to reduce regulatory T cells. No severe adverse events occurred. Analysis of pretreatment samples of malignant pleural effusion and ascites confirmed the efficacy of Ad5/3-D24-GMCSF in transduction and cell killing. Evidence of biological activity of the virus was seen in 13/21 patients and 8/12 showed objective clinical benefit as evaluated by radiology with Response Evaluation Criteria In Solid Tumors (RECIST) criteria. Antiadenoviral and antitumoral immune responses were elicited after treatment. Thus, Ad5/3-D24-GMCSF seems safe in treating cancer patients and promising signs of efficacy were seen.


Cancer Research | 2010

Oncolytic Adenovirus Coding for Granulocyte Macrophage Colony-Stimulating Factor Induces Antitumoral Immunity in Cancer Patients

Vincenzo Cerullo; Sari Pesonen; Iulia Diaconu; Sophie Escutenaire; Petteri Arstila; Matteo Ugolini; Petri Nokisalmi; Mari Raki; Leena Laasonen; Merja Särkioja; Maria Rajecki; Lotta Kangasniemi; Kilian Guse; Andreas Helminen; Laura Ahtiainen; Ari Ristimäki; Anne Räisänen-Sokolowski; Elina Haavisto; Minna Oksanen; Eerika Karli; Aila Karioja-Kallio; Sirkka-Liisa Holm; Mauri Kouri; Timo Joensuu; Anna Kanerva; Akseli Hemminki

Granulocyte macrophage colony-stimulating factor (GMCSF) can mediate antitumor effects by recruiting natural killer cells and by induction of tumor-specific cytotoxic T-cells through antigen-presenting cells. Oncolytic tumor cell-killing can produce a potent costimulatory danger signal and release of tumor epitopes for antigen-presenting cell sampling. Therefore, an oncolytic adenovirus coding for GMCSF was engineered and shown to induce tumor-specific immunity in an immunocompetent syngeneic hamster model. Subsequently, 20 patients with advanced solid tumors refractory to standard therapies were treated with Ad5-D24-GMCSF. Of the 16 radiologically evaluable patients, 2 had complete responses, 1 had a minor response, and 5 had disease stabilization. Responses were frequently seen in injected and noninjected tumors. Treatment was well tolerated and resulted in the induction of both tumor-specific and virus-specific immunity as measured by ELISPOT and pentamer analysis. This is the first time that oncolytic virus-mediated antitumor immunity has been shown in humans. Ad5-D24-GMCSF is promising for further clinical testing.


Clinical Cancer Research | 2010

Oncolytic Adenovirus ICOVIR-7 in Patients with Advanced and Refractory Solid Tumors

Petri Nokisalmi; Sari Pesonen; Sophie Escutenaire; Merja Särkioja; Mari Raki; Vincenzo Cerullo; Leena Laasonen; Ramon Alemany; Juan J. Rojas; Manel Cascallo; Kilian Guse; Maria Rajecki; Lotta Kangasniemi; Elina Haavisto; Aila Karioja-Kallio; Päivi Hannuksela; Minna Oksanen; Anna Kanerva; Timo Joensuu; Laura Ahtiainen; Akseli Hemminki

Purpose: Twenty-one patients with cancer were treated with a single round of oncolytic adenovirus ICOVIR-7. Experimental Design: ICOVIR-7 features an RGD-4C modification of the fiber HI-loop of serotype 5 adenovirus for enhanced entry into tumor cells. Tumor selectivity is mediated by an insulator, a modified E2F promoter, and a Rb-binding site deletion of E1A, whereas replication is optimized with E2F binding hairpins and a Kozak sequence. ICOVIR-7 doses ranged from 2 × 1010 to 1 × 1012 viral particles. All patients had advanced and metastatic solid tumors refractory to standard therapies. Results: ICOVIR-7 treatment was well tolerated with mild to moderate fever, fatigue, elevated liver transaminases, chills, and hyponatremia. One patient had grade 3 anemia but no other serious side effects were seen. At baseline, 9 of 21 of patients had neutralizing antibody titers against the ICOVIR-7 capsid. Treatment resulted in neutralizing antibody titer induction within 4 weeks in 16 of 18 patients. No elevations of serum proinflammatory cytokine levels were detected. Viral genomes were detected in the circulation in 18 of 21 of patients after injection and 7 of 15 of the samples were positive 2 to 4 weeks later suggesting viral replication. Conclusions: Overall, objective evidence of antitumor activity was seen in 9 of 17 evaluable patients. In radiological analyses, 5 of 12 evaluable patients had stabilization or reduction in tumor size. These consisted of one partial response, two minor responses and two cases of stable disease, all occurring in patients who had progressive disease before treatment. In summary, ICOVIR-7 treatment is apparently safe, resulting in anticancer activity, and is therefore promising for further clinical testing. Clin Cancer Res; 16(11); 3035–43. ©2010 AACR.


International Journal of Cancer | 2012

Integrin targeted oncolytic adenoviruses Ad5‐D24‐RGD and Ad5‐RGD‐D24‐GMCSF for treatment of patients with advanced chemotherapy refractory solid tumors

Sari Pesonen; Iulia Diaconu; Vincenzo Cerullo; Sophie Escutenaire; Mari Raki; Lotta Kangasniemi; Petri Nokisalmi; Gianpietro Dotti; Kilian Guse; Leena Laasonen; Kaarina Partanen; Eerika Karli; Elina Haavisto; Minna Oksanen; Aila Karioja-Kallio; Päivi Hannuksela; Sirkka Liisa Holm; Satu Kauppinen; Timo Joensuu; Anna Kanerva; Akseli Hemminki

The safety of oncolytic viruses for treatment of cancer has been shown in clinical trials while antitumor efficacy has often remained modest. As expression of the coxsackie‐adenovirus receptor may be variable in advanced tumors, we developed Ad5‐D24‐RGD, a p16/Rb pathway selective oncolytic adenovirus featuring RGD‐4C modification of the fiber. This allows viral entry through alpha‐v‐beta integrins frequently highly expressed in advanced tumors. Advanced tumors are often immunosuppressive which results in lack of tumor eradication despite abnormal epitopes being present. Granulocyte‐macrophage colony stimulating factor (GMCSF) is a potent activator of immune system with established antitumor properties. To stimulate antitumor immunity and break tumor associated immunotolerance, we constructed Ad5‐RGD‐D24‐GMCSF, featuring GMCSF controlled by the adenoviral E3 promoter. Preliminary safety of Ad5‐D24‐RGD and Ad5‐RGD‐D24‐GMCSF for treatment of human cancer was established. Treatments with Ad5‐D24‐RGD (N = 9) and Ad5‐RGD‐D24‐GMCSF (N = 7) were well tolerated. Typical side effects were grade 1‐2 fatigue, fever and injection site pain. 77% (10/13) of evaluable patients showed virus in circulation for at least 2 weeks. In 3 out of 6 evaluable patients, disease previously progressing stabilized after a single treatment with Ad5‐RGD‐D24‐GMCSF. In addition, 2/3 patients had stabilization or reduction in tumor marker levels. All patients treated with Ad5‐D24‐RGD showed disease progression in radiological analysis, although 3/6 had temporary reduction or stabilization of marker levels. Induction of tumor and adenovirus specific immunity was demonstrated with ELISPOT in Ad5‐RGD‐D24‐GMCSF treated patients. RGD modified oncolytic adenoviruses with or without GMCSF seem safe for further clinical development.


Clinical Cancer Research | 2006

Infectivity-Enhanced Adenoviruses Deliver Efficacy in Clinical Samples and Orthotopic Models of Disseminated Gastric Cancer

Lotta Kangasniemi; Tuula Kiviluoto; Anna Kanerva; Mari Raki; Tuuli Ranki; Merja Särkioja; Hongju Wu; Frank C. Marini; Krister Höckerstedt; Helena Isoniemi; Henrik Alfthan; Ulf-Håkan Stenman; David T. Curiel; Akseli Hemminki

Purpose: Metastatic gastric cancer remains a common and devastating disease without curative treatment. Recent proof-of-concept clinical trials have validated gene therapy with adenoviruses as an effective and safe modality for the treatment of cancer. However, expression of the primary coxsackie-adenovirus receptor is variable in advanced cancers, and therefore, the use of heterologous receptors could be advantageous. Experimental Design: Here, we used capsid-modified adenoviruses for increasing the transduction and subsequent antitumor efficacy. 5/3 chimeric viruses have a serotype 3 knob which allows binding to a receptor distinct from coxsackie-adenovirus receptor. The fiber of Ad5lucRGD is modified with an integrin-targeted motif. Polylysine motifs, pK7 and pK21, bind to heparan sulfates. Oncolytic adenoviruses replicate in and kill tumor cells selectively. Gastric cancer cell lines and fresh clinical samples from patients were infected with transductionally targeted viruses. Capsid-modified oncolytic adenoviruses were used in cell killing experiments. To test viral transduction and therapeutic efficacy in vivo, we developed orthotopic mouse models featuring i.p. disseminated human gastric cancer, which allowed the evaluation of biodistribution and antitumor efficacy in a system similar to humans. Results: Capsid modifications benefited gene transfer efficiency and cell killing in gastric cancer cell lines and clinical samples in vitro and in vivo. Modified oncolytic adenoviruses significantly increased the survival of mice with orthotopic gastric cancer. Conclusions: These preclinical data set the stage for the clinical evaluation of safety and efficacy in patients with disease refractory to current modalities.


Biomaterials | 2013

SPECT/CT imaging of radiolabeled cubosomes and hexosomes for potential theranostic applications

Christa Nilsson; Brianda Barrios-Lopez; Annukka Kallinen; Pasi Laurinmäki; Sarah J. Butcher; Mari Raki; Janne Weisell; Kim A. Bergström; Susan Weng Larsen; Jesper Østergaard; Claus Larsen; Arto Urtti; Anu J. Airaksinen; Anan Yaghmur

We have developed a highly efficient method for the radiolabeling of phytantriol (PHYT)/oleic acid (OA)-based hexosomes based on the surface chelation of technetium-99m ((99m)Tc) to preformed hexosomes using the polyamine 1, 12-diamino-3, 6, 9-triazododecane (SpmTrien) as chelating agent. We also report on the unsuccessful labeling of cubosomes using the well-known chelating agent hexamethylpropyleneamine oxime (HMPAO). The (99m)Tc-labeled SpmTrien-hexosomes ((99m)Tc-SpmTrien-hexosomes) were synthesized with good radiolabeling (84%) and high radiochemical purity (>90%). The effect of radiolabeling on the internal nanostructure and the overall size of these aqueous dispersions was investigated by using synchrotron small angle X-ray scattering (SAXS), dynamic light scattering (DLS), and transmission electron cryo microscopy (cryo-TEM). Further, we show the utility of (99m)Tc-SpmTrien-hexosomes for the in vivo imaging of healthy mice using single photon emission computed tomography (SPECT) in combination with computed tomography (CT), i.e. SPECT/CT. SPECT/CT experiments of subcutaneously administered (99m)Tc-SpmTrien-hexosomes to the flank of mice showed a high stability in vivo allowing imaging of the distribution of the radiolabeled hexosomes for up to 24 h. These injected (99m)Tc-SpmTrien-hexosomes formed a deposit within the subcutaneous adipose tissue, displaying a high biodistribution of ≈ 343% injected dose/g tissue (%ID/g), with negligible uptake in other organs and tissues. The developed (99m)Tc labeling method for PHYT/OA-based hexosomes could further serve as a useful tool for investigating and imaging the in vivo performance of cubosomal and hexosomal drug nanocarriers.


Molecular Therapy | 2011

Induction of interferon pathways mediates in vivo resistance to oncolytic adenovirus.

Ilkka Liikanen; Vladia Monsurrò; Laura Ahtiainen; Mari Raki; Tanja Hakkarainen; Iulia Diaconu; Sophie Escutenaire; Otto Hemminki; João D. Dias; Vincenzo Cerullo; Anna Kanerva; Sari Pesonen; Daniela Marzioni; Marco Colombatti; Akseli Hemminki

Oncolytic adenoviruses are an emerging experimental approach for treatment of tumors refractory to available modalities. Although preclinical results have been promising, and clinical safety has been excellent, it is also apparent that tumors can become virus resistant. The resistance mechanisms acquired by advanced tumors against conventional therapies are increasingly well understood, which has allowed development of countermeasures. To study this in the context of oncolytic adenovirus, we developed two in vivo models of acquired resistance, where initially sensitive tumors eventually gain resistance and relapse. These models were used to investigate the phenomenon on RNA and protein levels using two types of analysis of microarray data, quantitative reverse transcriptase-polymerase chain reaction and immunohistochemistry. Interferon (IFN) signaling pathways were found upregulated and Myxovirus resistance protein A (MxA) expression was identified as a marker correlating with resistance, while transplantation experiments suggested a role for tumor stroma in maintaining resistance. Furthermore, pathway analysis suggested potential therapeutic targets in oncolytic adenovirus-resistant cells. Improved understanding of the antiviral phenotype causing tumor recurrence is of key importance in order to improve treatment of advanced tumors with oncolytic adenoviruses. Given the similarities between mechanisms of action, this finding might be relevant for other oncolytic viruses as well.


PLOS ONE | 2012

Pre-Targeting and Direct Immunotargeting of Liposomal Drug Carriers to Ovarian Carcinoma

Julia Lehtinen; Mari Raki; Kim A. Bergström; Päivi Uutela; Katariina Lehtinen; Annukka Hiltunen; Jere Pikkarainen; Huamin Liang; Sari Pitkänen; Ann-Marie Määttä; Raimo A. Ketola; Marjo Yliperttula; Thomas Wirth; Arto Urtti

Background Epidermal growth factor receptor (EGFR) is overexpressed in many solid tumor types, such as ovarian carcinoma. Immunoliposome based drug targeting has shown promising results in drug delivery to the tumors. However, the ratio of tumor-to-normal tissue concentrations should be increased to minimize the adverse effects of cytostatic drugs. Methodology/Principal Findings We studied the EGFR-targeted doxorubicin immunoliposomes using pre-targeting and local intraperitoneal (i.p.) administration of the liposomes. This approach was used to increase drug delivery to tumors as compared to direct intravenous (i.v.) administration of liposomes. EGFR antibodies were attached on the surface of PEG coated liposomes using biotin-neutravidin binding. Receptor mediated cellular uptake and cytotoxic efficacy of EGFR-targeted liposomes were investigated in human ovarian adenocarcinoma (SKOV-3 and SKOV3.ip1) cells. In vivo distribution of the liposomes in mice was explored using direct and pre-targeting approaches and SPECT/CT imaging. Targeted liposomes showed efficient and specific receptor-mediated binding to ovarian carcinoma cells in vitro, but the difference in cytotoxicity between targeted and non-targeted liposomes remained small. The relatively low cytotoxic efficacy is probably due to insufficient doxorubicin release from the liposomes rather than lack of target binding. Tumor uptake of targeted liposomes in vivo was comparable to that of non-targeted liposomes after both direct and pre-targeting administration. For both EGFR-targeted and non-targeted liposomes, the i.p. administration increased liposome accumulation to the tumors compared to i.v. injections. Conclusions/Significance Intraperitoneal administration of liposomes may be a beneficial approach to treat the tumors in the abdominal cavity. The i.p. pre-targeting method warrants further studies as a potential approach in cancer therapy.


Journal of Gene Medicine | 2011

Switching the fiber knob of oncolytic adenoviruses to avoid neutralizing antibodies in human cancer patients

Mari Raki; Merja Särkioja; Sophie Escutenaire; Lotta Kangasniemi; Elina Haavisto; Anna Kanerva; Vincenzo Cerullo; Timo Joensuu; Minna Oksanen; Sari Pesonen; Akseli Hemminki

Oncolytic adenoviruses are an attractive strategy for treating cancers resistant to conventional treatments. However, their systemic utility could be limited as a result of the high prevalence of pre‐existing immunity towards the vector. Furthermore, neutralizing antibodies (NAbs) may prevent successful intravenous readministration of the same agent. Previous preclinical reports indicate that the NAb response can be partially overcome by modifying the adenoviral fiber knob. However, to date, this strategy has not been evaluated in human patients.


Molecular Therapy | 2012

Verapamil Results in Increased Blood Levels of Oncolytic Adenovirus in Treatment of Patients With Advanced Cancer

Anniina Koski; Mari Raki; Petri Nokisalmi; Ilkka Liikanen; Lotta Kangasniemi; Timo Joensuu; Anna Kanerva; Sari Pesonen; Ramon Alemany; Akseli Hemminki

Calcium channel blockers including verapamil have been proposed to enhance release and antitumor efficacy of oncolytic adenoviruses in preclinical studies but this has not been studied in humans before. Here, we studied if verapamil leads to increased replication of oncolytic adenovirus in cancer patients, as measured by release of virions from tumor cells into the systemic circulation. The study was conducted as a matched case-control study of advanced cancer patients treated with oncolytic adenoviruses with or without verapamil. We observed that verapamil increased mean virus titers present in blood after treatment (P < 0.05). The frequency or severity of adverse events was not increased, nor were cytokine responses or neutralizing antibody levels different between groups. Signs of possible treatment-related clinical benefits were observed in both groups, but there was no significant difference in responses or survival. Thus, our data suggests that the combination of verapamil with oncolytic adenoviruses is safe and well tolerated. Moreover, verapamil treatment seems to result in higher virus titers in blood, indicating enhanced overall replication in tumors. A randomized trial is needed to confirm these findings and to study if enhanced replication results in benefits to patients.

Collaboration


Dive into the Mari Raki'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

Tuuli Ranki

University of Helsinki

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Timo Joensuu

Helsinki University Central Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge