Network


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

Hotspot


Dive into the research topics where Tuomo Alanko is active.

Publication


Featured researches published by Tuomo Alanko.


Journal of Clinical Oncology | 2009

Fluorouracil, Epirubicin, and Cyclophosphamide With Either Docetaxel or Vinorelbine, With or Without Trastuzumab, As Adjuvant Treatments of Breast Cancer: Final Results of the FinHer Trial

Heikki Joensuu; Petri Bono; Vesa Kataja; Tuomo Alanko; Riitta Kokko; Raija Asola; Taina Turpeenniemi-Hujanen; Sirkku Jyrkkiö; Kari Möykkynen; Leena Helle; Seija Ingalsuo; Marjo Pajunen; Mauri Huusko; Tapio Salminen; Päivi Auvinen; Hannu Leinonen; Mika Leinonen; Jorma Isola; Pirkko-Liisa Kellokumpu-Lehtinen

PURPOSE Docetaxel has not been compared with vinorelbine as adjuvant treatment of early breast cancer. Efficacy and long-term safety of a short course of adjuvant trastuzumab administered concomitantly with chemotherapy for human epidermal growth factor receptor 2 (HER2) -positive cancer are unknown. PATIENTS AND METHODS One thousand ten women with axillary node-positive or high-risk node-negative breast cancer were randomly assigned to receive three cycles of docetaxel or vinorelbine, followed in both groups by three cycles of fluorouracil, epirubicin, and cyclophosphamide (FEC). Women with HER2-positive cancer (n = 232) were further assigned to either receive or not receive trastuzumab for 9 weeks with docetaxel or vinorelbine. The median follow-up time was 62 months after random assignment. RESULTS Women assigned to docetaxel had better distant disease-free survival (DDFS) than those assigned to vinorelbine (hazard ratio [HR] = 0.66; 95% CI, 0.49 to 0.91; P = .010). In the subgroup of HER2-positive disease, patients treated with trastuzumab tended to have better DDFS than those treated with chemotherapy only (HR = 0.65; 95% CI, 0.38 to 1.12; P = .12; with adjustment for presence of axillary nodal metastases, HR = 0.57; P = .047). In exploratory analyses, docetaxel, trastuzumab, and FEC improved DDFS compared with docetaxel plus FEC (HR = 0.32; P = .029) and vinorelbine, trastuzumab, and FEC (HR = 0.31; P = .020). The median left ventricular ejection fraction of trastuzumab-treated patients remained unaltered during the 5-year follow-up; only one woman treated with trastuzumab was diagnosed with a heart failure. CONCLUSION Adjuvant treatment with docetaxel improves DDFS compared with vinorelbine. A brief course of trastuzumab administered concomitantly with docetaxel is safe and effective and warrants further evaluation.


British Journal of Cancer | 2011

Hypertension and overall survival in metastatic colorectal cancer patients treated with bevacizumab-containing chemotherapy

Pia Österlund; Leena-Maija Soveri; Helena Isoniemi; Tuija Poussa; Tuomo Alanko; Petri Bono

Background:Hypertension (HTN) is a common toxicity of anti-VEGF (vascular endothelial growth factor) antibody treatment. It may be a marker of VEGF signalling pathway inhibition and therefore represent a cancer biomarker in metastatic colorectal cancer (mCRC) patients treated with chemotherapy and bevacizumab.Methods:A total of 101 consecutive patients with mCRC were treated with standard chemotherapy combined with bevacizumab at dose of 2.5 mg kg−1 per week in a single centre. The median follow-up time of the patients alive was 64 months. Blood pressure was measured before each bevacizumab infusion, and HTN was graded according to common toxicity criteria for adverse events version 3.0.Results:Overall, 57 patients (56%) developed ⩾grade 1 HTN (median blood pressure 168/97 mm Hg), whereas 44 (44%) remained normotensive when treated with bevacizumab-containing chemotherapy regimen. Overall response rate was higher among patients with HTN (30 vs 20%; P=0.025). Hypertension was associated with improved progression-free survival (10.5 vs 5.3 months; P=0.008) and overall survival (25.8 vs 11.7 months; P<0.001), and development of HTN within 3 months had an independent, prognostic influence in a multivariate landmark survival analysis together with other known mCRC prognostic factors (P=0.007). There was no association between HTN and development of thromboembolic complications.Conclusion:Hypertension may predict outcome of bevacizumab-containing chemotherapy in mCRC. These data require confirmation in prospective studies including pharmacodynamic and pharmacokinetic analyses.


Cancer Research | 2012

Oncolytic Immunotherapy of Advanced Solid Tumors with a CD40L-Expressing Replicating Adenovirus: Assessment of Safety and Immunologic Responses in Patients

Sari Pesonen; Iulia Diaconu; Lotta Kangasniemi; Tuuli Ranki; Anna Kanerva; S Pesonen; Ulrike Gerdemann; Ann M. Leen; Kalevi Kairemo; Minna Oksanen; Elina Haavisto; Sirkka-Liisa Holm; Aila Karioja-Kallio; Satu Kauppinen; Kaarina Partanen; Leena Laasonen; T. Joensuu; Tuomo Alanko; Vincenzo Cerullo; Akseli Hemminki

The immunosuppressive environment of advanced tumors is a primary obstacle to the efficacy of immunostimulatory and vaccine approaches. Here, we report an approach to arm an oncolytic virus with CD40 ligand (CD40L) to stimulate beneficial immunologic responses in patients. A double-targeted chimeric adenovirus controlled by the hTERT promoter and expressing CD40L (CGTG-401) was constructed and nine patients with progressing advanced solid tumors refractory to standard therapies were treated intratumorally. No serious adverse events resulting in patient hospitalization occurred. Moderate or no increases in neutralizing antibodies were seen, suggesting effective Th1 immunologic effects. An assessment of the blood levels of virus indicated 17.5% of the samples (n = 40) were positive at a low level early after treatment, but not thereafter. In contrast, high levels of virus, CD40L, and RANTES were documented locally at the tumor. Peripheral blood mononuclear cells were analyzed by IFN-γ ELISPOT analysis and induction of both survivin-specific and adenovirus-specific T cells was seen. Antitumor T-cell responses were even more pronounced when assessed by intracellular cytokine staining after stimulation with tumor type-specific peptide pools. Of the evaluable patients, 83% displayed disease control at 3 months and in both cases in which treatment was continued the effect was sustained for at least 8 months. Injected and noninjected lesions responded identically. Together, these findings support further clinical evaluation of CGTG-401.


Clinical Cancer Research | 2013

Antiviral and Antitumor T-cell Immunity in Patients Treated with GM-CSF–Coding Oncolytic Adenovirus

Anna Kanerva; Petri Nokisalmi; Iulia Diaconu; Anniina Koski; Vincenzo Cerullo; Ilkka Liikanen; Siri Tähtinen; Minna Oksanen; Raita Heiskanen; S Pesonen; T. Joensuu; Tuomo Alanko; Kaarina Partanen; Leena Laasonen; Kalevi Kairemo; Sari Pesonen; Lotta Kangasniemi; Akseli Hemminki

Purpose: Multiple injections of oncolytic adenovirus could enhance immunologic response. In the first part of this article, the focus was on immunologic aspects. Sixty patients previously naïve to oncolytic virus and who had white blood cells available were treated. Thirty-nine of 60 were assessed after a single virus administration, whereas 21 of 60 received a “serial treatment” consisting of three injections within 10 weeks. In the second part, we focused on 115 patients treated with a granulocyte macrophage colony-stimulating factor (GM–CSF)–coding capsid chimeric adenovirus, CGTG-102. Results: Following serial treatment, both increase and decrease in antitumor T cells in blood were seen more frequently, findings which are compatible with induction of T-cell immunity and trafficking of T cells to tumors, respectively. Safety was good in both groups. In 115 patients treated with CGTG-102 (Ad5/3-D24-GMCSF), median overall survival was 111 days following single and 277 days after serial treatment in nonrandomized comparison. Switching the virus capsid for avoiding neutralizing antibodies in a serial treatment featuring three different viruses did not impact safety or efficacy. A correlation between antiviral and antitumor T cells was seen (P = 0.001), suggesting that viral oncolysis can result in epitope spreading and breaking of tumor-associated immunologic tolerance. Alternatively, some patients may be more susceptible to induction of T-cell immunity and/or trafficking. Conclusions: These results provide the first human data linking antiviral immunity with antitumor immunity, implying that oncolytic viruses could have an important role in cancer immunotherapy. Clin Cancer Res; 19(10); 2734–44. ©2013 AACR.


OncoImmunology | 2015

Repeated intratumoral administration of ONCOS-102 leads to systemic antitumor CD8+ T-cell response and robust cellular and transcriptional immune activation at tumor site in a patient with ovarian cancer

Lotta Vassilev; Tuuli Ranki; T. Joensuu; Elke Jäger; Julia Karbach; Claudia Wahle; Kaarina Partanen; Kalevi Kairemo; Tuomo Alanko; Riku Turkki; Nina Linder; Johan Lundin; Ari Ristimäki; Matti Kankainen; Akseli Hemminki; Charlotta Backman; Kasper Dienel; M von Euler; Elina Haavisto; Tiina Hakonen; Juuso Juhila; Magnus Jaderberg; Antti Vuolanto; S Pesonen

Adenoviruses are excellent immunotherapeutic agents with a unique ability to prime and boost immune responses. Recombinant adenoviruses cause immunogenic cancer cell death and subsequent release of tumor antigens for antigen presenting cells, resulting in the priming of potent tumor-specific immunity. This effect may be further enhanced by immune-stimulating transgenes expressed by the virus. We report a case of a 38-year-old female with Stage 3 metastatic micropapillary serous carcinoma of the ovary. She was treated in a Phase I study with a granulocyte-macrophage colony stimulating factor (GMCSF)-expressing oncolytic adenovirus, Ad5/3-D24-GMCSF (ONCOS-102). The treatment resulted in progressive infiltration of CD8+ lymphocytes into the tumor and concomitant systemic induction of several tumor-specific CD8+ T-cell populations. The patient was alive at the latest follow up more than 20 months after initiation of the study.


OncoImmunology | 2014

Local treatment of a pleural mesothelioma tumor with ONCOS-102 induces a systemic antitumor CD8 + T-cell response, prominent infiltration of CD8 + lymphocytes and Th1 type polarization

Tuuli Ranki; Timo Joensuu; Elke Jäger; Julia Karbach; Claudia Wahle; Kalevi Kairemo; Tuomo Alanko; Kaarina Partanen; Riku Turkki; Nina Linder; Johan Lundin; Ari Ristimäki; Matti Kankainen; Akseli Hemminki; Charlotta Backman; Kasper Dienel; Mikael von Euler; Elina Haavisto; Tiina Hakonen; Juuso Juhila; Magnus Jaderberg; Lotta Vassilev; Antti Vuolanto; Sari Pesonen

Late stage cancer is often associated with reduced immune recognition and a highly immunosuppressive tumor microenvironment. The presence of tumor infiltrating lymphocytes (TILs) and specific gene-signatures prior to treatment are linked to good prognosis, while the opposite is true for extensive immunosuppression. The use of adenoviruses as cancer vaccines is a form of active immunotherapy to initialise a tumor-specific immune response that targets the patients unique tumor antigen repertoire. We report a case of a 68-year-old male with asbestos-related malignant pleural mesothelioma who was treated in a Phase I study with a granulocyte-macrophage colony‑stimulating factor (GM-CSF)-expressing oncolytic adenovirus, Ad5/3-D24-GMCSF (ONCOS-102). The treatment resulted in prominent infiltration of CD8+ lymphocytes to tumor, marked induction of systemic antitumor CD8+ T-cells and induction of Th1-type polarization in the tumor. These results indicate that ONCOS-102 treatment sensitizes tumors to other immunotherapies by inducing a T-cell positive phenotype to an initially T-cell negative tumor.


Digestive Diseases and Sciences | 1998

Induction of proliferation in isolated guinea pig gastric epithelium during restitution after superficial injury

Arun Bhowmik; H. Paimela; Teemu Joutsi; Tuomo Alanko; Timo Paavonen; Olli Saksela; Eero Kivilaakso

Immediate repair of the gastrointestinalepithelium after superficial injury is calledrestitution. It is based on the migration of thesurviving mucoid neck cells over the area of injury. Theinvolvement of growth factors in the process has beenrecently documented. They are known to enhance theprocess (ie, EGF, FGF, TGF-β) and to activate thebasolateral Na+-H+-antiport (EGF).They may exert their effect by activating intracellular tyrosinekinases or by inducing chemotaxis. Yet, their precisemechanism of action in the process is unknown. The aimof the present study was to investigate the effect of modulation of the signal transductionpathway on the occurrence of proliferative mucoid neckand foveolar cells in guinea pig gastric epithelium.Therefore guinea pig gastric epithelium was mounted in Ussing chambers in vitro and perfused 4 hrafter superficial injury with 1.25 M NaCl. The potentialdifference over the epithelium and tissue resistancewere recorded simultaneously. The tissue was exposed either to cycloheximide, genistein, or to4-phorbol myristate 13-acetate (PMA) during the 4-hrrecovery, and the expression of proliferative cells wasassessed by staining the tissue for proliferative cells (Ki-67). The mean proliferative index oftissues subjected to NaCl injury was significantlyhigher than that of uninjured control tissues after 4 hrof restitution. Inhibition of the signaling pathway with genistein decreased the proliferative indexsignificantly, while its stimulation with phorbolmyristate increased it. Both electrophysiologic andmorphologic restitution were sensitive to genistein, but not to PMA or cycloheximide. Superficialepithelial injury results in a significantly increasedoccurrence of proliferative cells in isolated guinea piggastric epithelium. This endogenous activation of the tissue is sensitive to inhibition bytyrosine kinases and to stimulation by protein kinases.Electrophysiologic and morphologic recovery are alsoaffected by the modulation of the signaling pathway. This suggests that it is involved in theimmediate repair process.


Acta Oncologica | 2015

VMAT technique enables concomitant radiotherapy of prostate cancer and pelvic bone metastases

Timo Kiljunen; Aki Kangasmäki; Aili Aaltonen; Kalevi Kairemo; Kaarina Partanen; Greetta Joensuu; Tuomo Alanko; Leila Vaalavirta; Timo Joensuu

Abstract Background. Prostate cancer (PCa) patients with metastatic disease often suffer from skeletal pain and urinary retention impairing their quality of life. Prophylactic radiotherapy to bone metastases planned concomitantly with primary PCa radiotherapy could enable more precise control of combined dose in healthy tissues when compared to sequential palliative treatment. Materials and methods. Volumetric modulated arc therapy (VMAT) was planned for 14 PCa patients with primary bone metastases. The bone planning target volume (PTVbone) was contoured together with the PTVs of prostate (pr), pelvic lymph nodes (ln) and seminal vesicles (sv). Another virtual plan was calculated excluding PTVbone for dose volume histogram (DVH) comparison. DVHs were additionally compared to a set of actual VMAT treatment plans of a control cohort of 13 high risk PCa patients treated with PTVpr, PTVsv and PTVln. The prescribed doses varied between 42 and 76 Gy for PTVbone. Results. Recommended healthy tissue tolerances (Quantec) were not exceeded except for one patients rectum V50Gy value. Rectum doses did not increase significantly due to the inclusion of PTVbone. For bladder, there was a slight increase for V65Gy and V50Gy (2.7% and 7.4%). The DVHs of metastatic and non-metastatic patients were comparable. There were no differences in the PTVpr DVH parameters, while mean PTVln dose increased by 3.7 Gy–4.4 Gy due to the increased treatment volume related to PTVbone. All side effects were < grade 3 during the mean follow-up duration of 25 months. Conclusions. VMAT offers a good optimization tool for adding extra PTVs to the radiotherapy plan. Radiotherapy of bone metastases concomitantly with irradiation of the primary prostate tumor is a safe and well-tolerated approach and deserves to be studied in a randomized setting.


Journal for ImmunoTherapy of Cancer | 2014

Gene expression analysis of tumors demonstrates an induction of Th1 type immune response following intratumoral administration of ONCOS-102 in refractory solid tumor patients

Mamun Majumder; Ashwini Kumar; Caroline Heckman; Matti Kankainen; Sari Pesonen; Elke Jäger; Julia Karbach; Timo Joensuu; Kalevi Kairemo; Kaarina Partanen; Tuomo Alanko; Akseli Hemminki; Charlotta Backman; Kasper Dienel; Mikael von Euler; Tiina Hakonen; Juuso Juhila; Tuuli Ranki; Lotta Vassilev; Antti Vuolanto; Magnus Jaderberg

Advanced tumors are often immunosuppressive. Intratumoral administration of adenovirus activates Toll-like receptor signalling leading to production of pro-inflammatory cytokines and activation of the innate immune system. Oncolytic adenovirus causes immunogenic cancer cell death and creates a danger signal at tumors, thus undermining immunological tolerance towards tumors. The release of tumor antigens from dying cancer cells results in priming of a potent anti-tumor immune response. This effect may be enhanced by the local production of immunostimulatory molecules coded by the virus. We present results of gene expression analysis of tumors from a Phase I study with ONCOS-102, an oncolytic adenovirus coding for GMCSF, in 12 patients with refractory solid tumors. A total of 9 intratumoral injections of ONCOS-102 were given to each patient. Biopsies were collected at baseline and 1 and 2 months after treatment initiation. RNA was extracted from fresh-frozen biopsies using standard methods. Gene expression profiling was carried out using the Illumina BeadChip platform. Data was normalized by quantile method, probes presenting the same genes were averaged, and the batch effects were adjusted using ComBat pipeline, as implemented in Chipster (http://chipster.csc.fi/). Finally, log2 fold-changes were computed by subtracting baseline data from after treatment data. Network and pathway analyses were conducted through the use of IPA (Ingenuity ®Systems, http://www.ingenuity.com). A cutoff value of 2-fold expression change was used to filter differentially expressed genes and run core analysis to identify underlying signaling networks and deregulated molecules. A significant enrichment of genes involved in immune cell trafficking, inflammatory response and antigen presentation were detected post treatment. A mesothelioma patient showed a prominent post-treatment induction of MAGE3-specific CD8+ T-cells in peripheral blood in IFNγ ELISPOT. Furthermore, a late decrease in metabolic activity was observed in PET imaging 7.5 months after treatment initiation during the follow-up period, measured as a 47% decrease in total lesion glycolysis in comparison to the imaging at 6 months. In the same patient, upstream regulators driving differentially expressed genes detected in the post treatment biopsy included cytokines (INFG, TNF, IL1B, CCL2, CXCL10, IL-17A, CD40LG), enzymes (FN1, NOS2, PTGS2, PARP9), growth factors (BMP2, LEP), kinases (CHUK, CRKL, IKBKB, IKBKG, ITK, STK11, SYK), transcriptional regulators (IRF1, NFATC2, NFKBIA, STAT1, STAT3, ZEB1, RELA), and transmembrane receptors (B2M, CD40, IL6R, TLR2-4, TNFSF1B). Likely, these events collectively induced a higher CD8+ mediated cytotoxic T cell response (GZMB, PRF1) post treatment. Detailed analysis per patient will be presented at the meeting.


Journal for ImmunoTherapy of Cancer | 2015

Local immunotherapy with ONCOS-102 shapes harmful tumor associated CD68+ macrophages to become beneficial cells that correlate with increased overall survival

Sari Pesonen; Johan Lundin; Nina Linder; Riku Turkki; Ari Ristimäki; Timo Joensuu; Kalevi Kairemo; Kaarina Partanen; Tuomo Alanko; Elke Jäger; Julia Karbach; Claudia Wahle; Akseli Hemminki; Charlotta Backman; Mikael von Euler; Tiina Hakonen; Tuuli Ranki; Antti Vuolanto; Magnus Jaderberg; Dmitry Zamarin

With the increasing excitement around new immunotherapeutic approaches, there has been a shift in the way viral cancer therapy is regarded from providing mainly oncolysis towards being a cancer immunotherapy. Adenoviruses have a unique ability to prime and boost immune responses. GM-CSF coding adenovirus ONCOS-102 causes immunogenic cancer cell death whereupon tumor antigens are presented into the immunogenic environment. ONCOS-102 has been previously shown to initiate CD8+ T cell responses against tumor-derived antigens in chemotherapy refractory cancer patients. A total of 12 cancer patients were treated with repeated intratumoral injections of ONCOS-102 in a Phase I study. Sequential biopsies were collected at baseline and 1 and 2 months after treatment initiation and analyzed for the presence of immune cells by immunohistochemistry (IHC) in digitally scanned samples. Readout of the expression levels for innate immune cells (CD68, CD163, CD11c), T cells (CD3, CD4, CD8), and B cells (CD19) was performed in tumorous regions by the use of an image analysis algorithm based on color deconvolution and segmentation of the IHC stained cells. In an exploratory analysis, correlation between absolute expression levels of different immune cell markers in tumors and overall survival (OS) was assessed by Spearman´s rank correlation analysis. At baseline, the absolute expression level of macrophage marker CD68 negatively correlated with OS (correlation coefficient (r)= -0.59, p=0.04), suggesting that tumor-associated macrophages (TAMs) in untreated tumors were tumorigenic. No correlation between other immune cell markers and OS was seen at baseline. 11/12 patients showed a post-treatment increase in tumor-infiltrating innate and adaptive immune cells, with the most prominent increase seen in CD8+ cells. In contrast to baseline, post-treatment samples showed a positive correlation between the expression level of CD68+ cells and OS (r=0.71, p=0.01), suggesting that CD68+ macrophages that were attracted into tumors after ONCOS-102 injection displayed different functionality than TAMs present prior to treatment. Furthermore, absolute expression levels of T cell markers CD3 (r=0.74, p=0.006), CD4 (r=0.76, p=0.004), and CD8 (r=0.73, p=0.007) in post-treatment biopsies all positively correlated with OS. To summarize, ONCOS-102 induced infiltration of CD68+ macrophages and T cells which was associated with increased OS, while CD68+ TAMs pre-treatment was associated with shortened OS. This suggest that local immunotherapy with ONCOS-102 has the potential to activate immunologically silent tumors and reduce local immune suppression in advanced tumors.

Collaboration


Dive into the Tuomo Alanko's collaboration.

Top Co-Authors

Avatar

Kalevi Kairemo

Helsinki University Central Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
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

Tuuli Ranki

University of Helsinki

View shared research outputs
Top Co-Authors

Avatar

Elke Jäger

Nara Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge