353 research outputs found

    Improving adenovirus-based immunotherapies for treatment of solid tumors

    Get PDF
    New treatment modalities are needed for patients with advanced cancer, who have undergone several unsuccessful pretreatments. Cancer immunotherapy has emerged as a promising field of medicine with the potential to induce durable responses in these patients. Within the immunotherapy field, a diverse set of approaches have been employed, all of which aim at combating tumors with the cells of the immune system. Oncolytic immunotherapy encompasses the use of genetically engineered viruses to specifically kill (lyse) tumor cells and, importantly, to induce an antitumor immune response in the process. Oncolytic adenoviruses in particular possess an excellent safety profile and can be armed with immunostimulatory transgenes for the enhancement of antitumor immunity. In the first part of the thesis, oncolytic adenovirus armed with granulocyte-macrophage colony-stimulating factor (GMCSF) was used together with the chemotherapeutic agents doxorubicin and ifosfamide to treat soft-tissue sarcoma (STS) in an adenovirus-permissive Syrian hamster model. The combination treatment was highly effective against syngeneic hamster leiomyosarcoma tumors in vivo, with indications that adenovirus replication was improved in the presence of doxorubicin and that oncolytic adenovirus/chemotherapy combination induced immunogenic cell death (ICD) of tumor cells. Tumor-infiltrating lymphocytes (TIL) from syngeneic Syrian hamster tumors were cultured, characterized and used therapeutically with oncolytic adenovirus in the second part of the thesis. Co-treatment of pancreatic cancer tumors with adoptive transfer of pancreatic cancer derived TIL and oncolytic adenovirus resulted in improved antitumor efficacy when compared with either monotherapy. In the third part, non-replicating adenovirus vectors coding for the murine cytokines tumor necrosis factor alpha (TNFa) and interleukin 2 (IL-2) were constructed and used in combination with adoptive transfer of tumor-specific T-cell receptor engineered (TCR) T-cells for the treatment of immunosuppressive melanoma. This combination showed significant antitumor efficacy over single agent treatments. Mechanistic studies revealed that intratumoral virus injections induce trafficking of adoptively transferred T-cells to tumors. Furthermore, the cytokine-coding adenoviruses caused favorable alterations in the tumor microenvironment. In the final part of the thesis, oncolytic adenoviruses coding for human versions of TNFa and IL-2 were used with hamster TIL to successfully treat pancreatic cancer tumors. In fact, virus injections were capable of eliminating most tumors when combined TIL transfer, and protected cured hamsters from tumor rechallenge. From a safety perspective it is noteworthy that virus-mediated cytokine production was restricted to tumors, as negligible levels of cytokines were observed in the sera of intratumorally injected animals. In conclusion, the combinatorial approach studied in the preclinical setting here represents a rational and effective solution for the treatment of advanced solid tumors, warranting the clinical translation of adenovirus-based immunotherapy combined with other immunotherapies.Levinneen syövän uusista hoitokeinoista lupaavimpia on syövän immunoterapia. Tässä terapiamuodossa elimistön immuunisolut aktivoidaan hyökkäämään syöpäkasvaimia vastaan. Onkolyyttinen (syöpäsoluja hajottava) immunoterapia käsittää geneettisesti muokattujen virusten käytön tähän tarkoitukseen. Erityisesti adenovirukset ovat lupaavia, sillä niiden turvallisuusprofiili on erinomainen ja niiden genomiin voidaan lisätä immuunivastetta stimuloivia transgeenejä. Väitöskirjatutkimuksen ensimmäisessä osatyössä tutkittiin kemoterapian ja onkolyyttisen adenoviruksen yhdistelmiä pehmytkudossarkoomien hoitoon prekliinisissä eläinmalleissa. Yhdistelmähoidon havaittiin estävän tehokkaasti ihonalaisten syöpäkasvaimien kasvua eläimissä ilman vakavia sivuvaikutuksia. Lisääntyneen tehokkuuden taustalla havaittiin olevan kaksi mekanismia: immunogeenisen solukuoleman ja adenoviruksen replikaation tehostuminen kemoterapian läsnäollessa. Toisessa osatyössä tutkittiin onkolyyttisen viroterapian ja adoptiivisen T-solusiirron yhdistämistä kultahamstereissa (Mesocricetus auratus). Ihmisen adenovirukset voivat replikoitua hamsterin kudoksissa (toisin kuin hiiren tai rotan), minkä vuoksi hamsteri on hyödyllinen prekliininen eläinmalli tutkittaessa onkolyyttisiä adenoviruksia. Hamsterin haimasyöpäkasvaimista eristetyt ja monistetut T-solut annettiin takaisin kasvaimia kantaneille hamstereille yhdessä adenovirushoidon kanssa; yhdistelmähoito oli tehokkaampi kuin T-soluhoito tai adenovirushoito yksinään. Melanooman hoitoa adenoviruksilla ja adoptiivisella T-solusiirrolla tutkittiin väitöskirjan kolmannessa osatyössä. Hiirimalleissa havaittiin, että immuunivastetta stimuloivia sytokiinejä (TNFa ja IL-2) koodaavat adenovirusvektorit tehostivat T-solusiirtoa kun viruksia injektoitiin melanoomakasvaimiin. Kuvantamiskokeissa todettiin, että radioaktiivisesti leimatut kasvainspesifiset T-solut kulkeutuivat tehokkaimmin kasvaimiin, joihin oli injektoitu molempia sytokiinejä koodaavia adenoviruksia. Lisäksi adenovirushoidon havaittiin muokkaavan kasvaimen mikroympäristöä T-solusiirteelle suotuisammaksi, toisin sanoen vähemmän immunosuppressiiviseksi. Neljännessä osatyössä rakennettiin ihmisen kudoksissa aktiivisia sytokiinejä koodaava onkolyyttinen adenovirus. Uusi virus tehosti T-soluhoitoa hamsterimallissa merkittävästi; yhdistelmähoito paransi suuren osan kasvaimia kantaneista hamstereista ja tuotti niille immuniteetin kasvaintyyppiä vastaan. Väitöskirjatyön tulokset tukevat adenovirushoidon yhdistämistä muihin syövän hoitomuotoihin. Tutkimukset luovat pohjan kliinisten kokeiden suunnitellulle potilaissa, joiden levinneeseen syöpään ei ole muita hoitokeinoja

    Youth work and climate change : A follow up on the 2019 Finland's Presidency EU Council debate and questions about future

    Get PDF
    A survey on the national youth policy representatives of the EU countries looked at the effects of youth climate activism on youth work and youth policy: what was its visibility, has it appeared on youth work agendas, how should the youth field react to it, what are the future plans of support and the relationship to EU climate policies? Results showed that climate change had started to appear on the youth work agenda, but not yet as a universally adopted priority. Young people were felt to have the right to strike for the climate and youth work and youth policy were to react to this concern, but responses were divided as to the significance of the global strikes. Almost 2/3 thought that mass demonstrations represent an emergent way of youth agency, while 1/3 felt they were only a singular phenomenon. Respondents were "hesitantly favourable" to perceive climate change as a youth field challenge. Also, not all respondents "totally agreed" to engage in collaboration with European Climate Pact or to utilise Erasmus+ to combat climate change. The study discusses whether youth climate activism signify a change of youth participation paradigm and the identity, resilience and agility of European youth policy and youth work to react to emerging challenges like climate change. The study should be replicated to follow the 'greening' of European youth policy and youth work

    Kiihtelysvaaran tapaus

    Get PDF
    Summary : The social meaning of alcohol - the Kiihtelysvaara case

    Nuoruus sosiaalisena sopimuksena - suomalaisen nuorisotutkimuksen virstanpylväs

    Get PDF
    Kirja-arvostelu: Nuorisokulttuureista kulttuuriseen nuoruuteen. Tommi Hoikkal

    Cytokine-Coding Oncolytic Adenovirus TILT-123 Is Safe, Selective, and Effective as a Single Agent and in Combination with Immune Checkpoint Inhibitor Anti-PD-1

    Get PDF
    Oncolytic viruses provide a biologically multi-faceted treatment option for patients who cannot be cured with currently available treatment options. We constructed an oncolytic adenovirus, TILT-123, to support T-cell therapies and immune checkpoint inhibitors in solid tumors. Adenoviruses are immunogenic by nature, are easy to produce in large quantities, and can carry relatively large transgenes. They are the most commonly used gene therapy vectors and are well tolerated in patients. TILT-123 expresses two potent cytokines, tumor necrosis factor alpha and interleukin-2, to stimulate especially the T-cell compartment in the tumor microenvironment. Before entering clinical studies, the safety and biodistribution of TILT-123 was studied in Syrian hamsters and in mice. The results show that TILT-123 is safe in animals as monotherapy and in combination with an immune checkpoint inhibitor anti-PD-1. The virus treatment induces acute changes in circulating immune cell compartments, but the levels return to normal by the middle of the treatment period. The virus is rapidly cleared from healthy tissues, and it does not cause damage to vital organs. The results support the initiation of a phase 1 dose-escalation trial, where melanoma patients receiving a tumor-infiltrating lymphocyte therapy are treated with TILT-123 (NCT04217473).Peer reviewe

    Adenovirus Coding for Interleukin-2 and Tumor Necrosis Factor Alpha Replaces Lymphodepleting Chemotherapy in Adoptive T Cell Therapy

    Get PDF
    Lymphodepleting preconditioning with high-dose chemotherapy is commonly used to increase the clinical efficacy of adoptive T cell therapy (ACT) strategies, however, with severe toxicity for patients. Conversely, oncolytic adenoviruses are safe and, when engineered to express interleukin-2 (IL-2) and tumor necrosis factor alpha (TNF-alpha), they can achieve antitumor immunomodulatory effects similar to lymphodepletion. Therefore, we compare the safety and efficacy of such adenoviruses with a cyclophosphamide-and fludarabine- containing lymphodepleting regimen in the setting of ACT. Human adenovirus (Ad5/3-E2F-D24-hTNF-alpha-IRES-hIL-2; TILT-123) replication was studied using a Syrian hamster pancreatic tumor model (HapT1) infused with tumor- infiltrating lymphocytes (TILs). Using the oncolytic virus instead of lymphodepletion resulted in superior efficacy and survival. Immune cells responsive to TNF-alpha IL-2 were studied using an immunocompetent mouse melanoma model (B16. OVA) infused with ovalbumin-specific T (OT-I) cells. Here, the adenovirus approach improved tumor control together with increased intratumoral Th1 cytokine levels and infiltration of CD8+ T cells and CD86+ dendritic cells. Similar to humans, lymphodepleting preconditioning caused severe cytopenias, systemic inflammation, and damage to vital organs. Toxicity was minimal in adenovirus- and OT-Itreated mice. These findings demonstrate that ACT can be effectively facilitated by cytokine-coding adenovirus without requiring lymphodepletion, a rationale being clinically investigated.Peer reviewe

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

    Get PDF
    Releasing the patient's 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.Peer reviewe
    corecore