55 research outputs found

    Applications of biomimetic nanoparticles in breast cancer as a blueprint for improved next-generation cervical cancer therapy

    Get PDF
    Nanomedicines are innovative and promising, but lack a convincing clinical presence. Thus, biomimetic nanoparticles (BMNPs) have been designed with functionalizations which structurally and/or functionally mimic the biological setting, endowing thereupon biological structure and functionality. These may be coated with biologically derived materials, but may also include artificial antigen-presenting cells and synthetic architectures. When applied in cancer theranostics, BMNPs show significant improvements over traditional drugs and similar non-biomimetic NPs, especially in terms of circulation time, tissue penetration, delivery, and lowered toxicity. These particles have achieved unprecedented outcomes through top-down synthesis methods (cell material to NP), which bypass complex bottom-up synthetic techniques attempting to mimic such complex and diverse biological components. Breast cancer has received much attention in this area, and as such, is studied in this paper as a template for how BMNPs could be applied in cervical cancer – an area with few BMNP applications and a dire need for efficacious and fertility-preserving therapies. This cancer remains an enormous burden globally, especially in developing countries. Being a virus-induced disease, biomimetic applications may be particularly promising, aligning with the emergence of biomimetic nanovaccines in recent years. Feasibility challenges remain within BMNPs: Extracting biological material for re-administration to patients could cause ethical debate, and the costs involved in preparing scaled up quantities of biomimetic NPs would be large. However, with a clearer understanding and tighter characterization of preparation methods and biological responses, BMNPs may add great value to the nanomedicine community.</p

    Elevated temperatures and longer durations improve the efficacy of oxaliplatin- and mitomycin C-based hyperthermic intraperitoneal chemotherapy in a confirmed rat model for peritoneal metastasis of colorectal cancer origin

    Get PDF
    Introduction: In patients with limited peritoneal metastasis (PM) originating from colorectal cancer, cytoreductive surgery (CRS) followed by hyperthermic intraperitoneal chemotherapy (HIPEC) is a potentially curative treatment option. This combined treatment modality using HIPEC with mitomycin C (MMC) for 90 minutes proved to be superior to systemic chemotherapy alone, but no benefit of adding HIPEC to CRS alone was shown using oxaliplatin-based HIPEC during 30 minutes. We investigated the impact of treatment temperature and duration as relevant HIPEC parameters for these two chemotherapeutic agents in representative preclinical models. The temperature- and duration- dependent efficacy for both oxaliplatin and MMC was evaluated in an in vitro setting and in a representative animal model. Methods: In 130 WAG/Rij rats, PM were established through i.p. injections of rat CC-531 colon carcinoma cells with a signature similar to the dominant treatment-resistant CMS4 type human colorectal PM. Tumor growth was monitored twice per week using ultrasound, and HIPEC was applied when most tumors were 4-6 mm. A semi-open four-inflow HIPEC setup was used to circulate oxaliplatin or MMC through the peritoneum for 30, 60 or 90 minutes with inflow temperatures of 38°C or 42°C to achieve temperatures in the peritoneum of 37°C or 41°C. Tumors, healthy tissue and blood were collected directly or 48 hours after treatment to assess the platinum uptake, level of apoptosis and proliferation and to determine the healthy tissue toxicity. Results: In vitro results show a temperature- and duration- dependent efficacy for both oxaliplatin and MMC in both CC-531 cells and organoids. Temperature distribution throughout the peritoneum of the rats was stable with normothermic and hyperthermic average temperatures in the peritoneum ranging from 36.95-37.63°C and 40.51-41.37°C, respectively. Treatments resulted in minimal body weight decrease (&lt;10%) and only 7/130 rats did not reach the endpoint of 48 hours after treatment. Conclusions: Both elevated temperatures and longer treatment duration resulted in a higher platinum uptake, significantly increased apoptosis and lower proliferation in PM tumor lesions, without enhanced normal tissue toxicity. Our results demonstrated that oxaliplatin- and MMC-based HIPEC procedures are both temperature- and duration-dependent in an in vivo tumor model.</p

    Enhancing synthetic lethality of PARP-inhibitor and cisplatin in BRCA-proficient tumour cells with hyperthermia

    Get PDF
    Background: Poly-(ADP-ribose)-polymerase1 (PARP1) is involved in repair of DNA single strand breaks. PARP1-inhibitors (PARP1-i) cause an accumulation of DNA double strand breaks, which are generally repaired by homologous recombination (HR). Therefore, cancer cells harboring HR deficiencies are exceptionally sensitive to PARP1-i. For patients with HR-proficient tumors, HR can be temporarily inhibited by hyperthermia, thereby inducing synthetic lethal conditions in every tumor type. Since cisplatin is successfully used combined with hyperthermia (thermochemotherapy), we investigated the effectiveness of combining PARP1-i with thermochemotherapy. Results: The in vitro data demonstrate a decreased in cell survival after addition of PARP1-i to thermochemotherapy, which can be explained by increased DNA damage induction and less DSB repair. These in vitro findings are in line with in vivo model, in which a decreased tumor growth is observed upon addition of PARP1-i. Materials and Methods: Survival of three HR-proficient cell lines after cisplatin, hyperthermia and/or PARP1-i was studied. Cell cycle analyses, quantification of γ-H2AX foci and apoptotic assays were performed to understand these survival data. The effects of treatments were further evaluated by monitoring tumor responses in an in vivo rat model. Conclusions: Our results in HR-proficient cell lines suggest that PARP1-i combined with thermochemotherapy can be a promising clinical approach for all tumors independent of HR status

    Sensitizing thermochemotherapy with a PARP1-inhibitor

    Get PDF
    Cis-diamminedichloroplatinum(II) (cisplatin, cDDP) is an effective chemotherapeutic agent that induces DNA double strand breaks (DSBs), primarily in replicating cells. Generally, such DSBs can be repaired by the classical or backup non-homologous end joining (c-NHEJ/b-NHEJ) or homologous recombination (HR). Therefore, inhibiting these pathways in cancer cells should enhance the efficiency of cDDP treatments. Indeed, inhibition of HR by hyperthermia (HT) sensitizes cancer cells to cDDP and in the Netherlands this combination is a standard treatment option for recurrent cervical cancer after previous radiotherapy. Additionally, cDDP has been demonstrated to disrupt c-NHEJ, which likely further increases the treatment efficacy. However, if one of these pathways is blocked, DSB repair functions can be sustained by the Poly-(ADP-ribose)-polymerase1 (PARP1)-dependent b-NHEJ. Therefore, disabling b-NHEJ should, in principle, further inhibit the repair of cDDP-induced DNA lesions and enhance the toxicity of thermochemotherapy. To explore this hypothesis, we treated a panel of cancer cell lines with HT, cDDP and a PARP1-i and measured various end-point relevant in cancer treatment. Our results demonstrate that PARP1-i does not considerably increase the efficacy of HT combined with standard, commonly used cDDP concentrations. However, in the presence of a PARP1-i, ten-fold lower concentration of cDDP can be used to induce similar cytotoxic effects. PARP1 inhibition may thus permit a substantial lowering of cDDP concentrations without diminishing treatment efficacy, potentially reducing systemic side effects

    Hyperthermia-based anti-cancer treatments

    No full text

    The role of hyperthermia in the treatment of locally advanced cervical cancer: a comprehensive review

    No full text
    Radiotherapy with cisplatin (chemoradiation) is the standard treatment for women with locally advanced cervical cancer. Radiotherapy with deep hyperthermia (thermoradiation) is a well established alternative, but is rarely offered as an alternative to chemoradiation, particularly for patients in whom cisplatin is contraindicated. The scope of this review is to provide an overview of the biological rationale of hyperthermia treatment delivery, including patient workflow, and the clinical effectiveness of hyperthermia as a radiosensitizer in the treatment of cervical cancer. Hyperthermia is especially effective in hypoxic and nutrient deprived areas of the tumor where radiotherapy is less effective. Its radiosensitizing effectiveness depends on the temperature level, duration of treatment, and the time interval between radiotherapy and hyperthermia. High quality hyperthermia treatment requires an experienced team, adequate online adaptive treatment planning, and is preferably performed using a phased array radiative locoregional hyperthermia device to achieve the optimal thermal dose effect. Hyperthermia is well tolerated and generally leads to only mild toxicity, such as patient discomfort. Patients in whom cisplatin is contraindicated should therefore be referred to a hyperthermia center for thermoradiation

    Effects of hyperthermia on DNA repair pathways: one treatment to inhibit them all

    No full text
    The currently available arsenal of anticancer modalities includes many DNA damaging agents that can kill malignant cells. However, efficient DNA repair mechanisms protect both healthy and cancer cells against the effects of treatment and contribute to the development of drug resistance. Therefore, anti-cancer treatments based on inflicting DNA damage can benefit from inhibition of DNA repair. Hyperthermia - treatment at elevated temperature - considerably affects DNA repair, among other cellular processes, and can thus sensitize (cancer) cells to DNA damaging agents. This effect has been known and clinically applied for many decades, but how heat inhibits DNA repair and which pathways are targeted has not been fully elucidated. In this review we attempt to summarize the known effects of hyperthermia on DNA repair pathways relevant in clinical treatment of cancer. Furthermore, we outline the relationships between the effects of heat on DNA repair and sensitization of cells to various DNA damaging agent

    The impact of the time interval between radiation and hyperthermia on clinical outcome in patients with locally advanced cervical cancer

    No full text
    The benefit of hyperthermia combined with radiotherapy is well-acknowledged for patients with locally advanced cervical cancer (LACC) (1-3). However, recently a discussion evolved on the optimal time interval between radiotherapy and hyperthermia. Kroesen et al. (4) recently reported a retrospective analysis of factors influencing clinical results of treatment with radiotherapy and hyperthermia in a large cohort of locally advanced cervical cancer patients (LACC) at ErasmusMC in Rotterdam. They concluded that there is no detrimental effect of prolonged intervals on clinical outcome within a time frame of 4 h between radiotherapy and hyperthermia. Kroesen et al. thereby explicitly dismissed the findings of Van Leeuwen et al. (5) in a smaller cohort of LACC patients treated at the Academic Medical Center (AMC) of the University of Amsterdam. In that study longer time intervals and lower tumor temperatures were both found to have a highly negative effect on in-field tumor control (time interval: P = 0.021, in multivariable analysis p = 0.007) and overall survival (idem: P = 0.015, in multivariable analysis p = 0.012), where it is important to note that the median time intervals between radiotherapy and hyperthermia were ∼60 and ∼90 min for the short and long time interval subgroups of patients, respectively
    • …
    corecore