Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 10.514
1.
Skin Res Technol ; 30(6): e13766, 2024 Jun.
Article En | MEDLINE | ID: mdl-38807440

INTRODUCTION: Severe acne breakouts often lead to atrophic acne scars, which affect millions of people worldwide and can significantly affect a person's self-confidence and self-image. Given the difficulty in treating atrophic acne scars, this study aims to investigate the efficacy of topical phenytoin in the treatment of atrophic acne scars. METHOD: This split face clinical trial on 25 patients between the ages of 18 and 40 involved the application of microneedling on one side of the face, with three sessions taking place over the course of a month. On the other side, a 1% phenytoin cream was administered three times daily for 1 week following the microneedling procedure. Baseline information was collected for all patients, and follow-up assessments were conducted during the treatment sessions and 2 months after the last session. The assessments included evaluating the number and area of pores and spots, determining scar severity, assessing patient satisfaction, and recording any potential complications. RESULTS: Among patients, 20 individuals (80%) were females, and the average age of the participants was 35.96 ± 9.23. In terms of the fine pore area, despite the fine pore count, both groups showed improvement over time (p: 0.03 vs. 0.06). Also, regarding large pore count and area, and the count and area of spots, both groups showed improvement over time (p: 0.001). However, there were no significant differences between the two groups (p > 0.05). On the other hand, when it comes to acne scar grade and patients' satisfaction, the phenytoin group outperformed the control group in all follow-up sessions and this difference was found to be significant (p: 0.001). It is worth noting that no complications were observed among any of the patients. CONCLUSION: It appears that combining phenytoin cream with microneedling has a more effective therapeutic outcome in enhancing atrophic acne scars, when compared to microneedling alone, and this method can be regarded as a viable alternative in treating these types of scars.


Acne Vulgaris , Cicatrix , Needles , Phenytoin , Humans , Female , Phenytoin/administration & dosage , Phenytoin/therapeutic use , Adult , Acne Vulgaris/complications , Acne Vulgaris/therapy , Acne Vulgaris/pathology , Male , Cicatrix/etiology , Cicatrix/pathology , Young Adult , Adolescent , Treatment Outcome , Patient Satisfaction , Administration, Cutaneous , Combined Modality Therapy/methods , Atrophy , Administration, Topical , Percutaneous Collagen Induction
2.
Sci Rep ; 14(1): 11569, 2024 05 21.
Article En | MEDLINE | ID: mdl-38773258

Combining radiation therapy with immunotherapy is a strategy to improve both treatments. The purpose of this study was to compare responses for two syngeneic head and neck cancer (HNC) tumor models in mice following X-ray or proton irradiation with or without immune checkpoint inhibition (ICI). MOC1 (immunogenic) and MOC2 (less immunogenic) tumors were inoculated in the right hind leg of each mouse (C57BL/6J, n = 398). Mice were injected with anti-PDL1 (10 mg/kg, twice weekly for 2 weeks), and tumors were treated with single-dose irradiation (5-30 Gy) with X-rays or protons. MOC2 tumors grew faster and were more radioresistant than MOC1 tumors, and all mice with MOC2 tumors developed metastases. Irradiation reduced the tumor volume in a dose-dependent manner. ICI alone reduced the tumor volume for MOC1 with 20% compared to controls, while no reduction was seen for MOC2. For MOC1, there was a clear treatment synergy when combining irradiation with ICI for radiation doses above 5 Gy and there was a tendency for X-rays being slightly more biologically effective compared to protons. For MOC2, there was a tendency of protons being more effective than X-rays, but both radiation types showed a small synergy when combined with ICI. Although the responses and magnitudes of the therapeutic effect varied, the optimal radiation dose for maximal synergy appeared to be in the order of 10-15 Gy, regardless of tumor model.


Immunotherapy , Proton Therapy , Animals , Mice , Proton Therapy/methods , Immunotherapy/methods , Mouth Neoplasms/radiotherapy , Mouth Neoplasms/therapy , Mouth Neoplasms/immunology , Mouth Neoplasms/pathology , Mice, Inbred C57BL , Cell Line, Tumor , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , X-Rays , Combined Modality Therapy/methods , X-Ray Therapy , Female , Disease Models, Animal
3.
Acta Neurochir (Wien) ; 166(1): 218, 2024 May 15.
Article En | MEDLINE | ID: mdl-38750340

PURPOSE: The aim of this study was to evaluate the treatment outcomes of neuroendoscopic cyst partial resection (ECPR) combined with stereotactic radiotherapy (SRT) for cystic craniopharyngiomas. METHODS: In this retrospective study, 22 craniopharyngioma patients undergoing ECPR combined with SRT were included. This combination therapy was indicated for suprasellar cystic craniopharyngiomas in patients whose pituitary function was preserved but would be difficult to preserve in direct surgery. The outcomes of combination therapy, including tumor control and postoperative visual and pituitary functions, were investigated. RESULTS: ECPR was safely performed, and cyst shrinkage was accomplished in all cases. After ECPR, visual function improved in 12 of 13 patients (92%) with visual field disturbance and did not deteriorate in any patients. Pituitary function was preserved in 14 patients (64%) and deteriorated in eight patients (36%) after ECPR. As a complication of ECPR, meningitis occurred because of a wound infection in one patient. In 18 of 22 patients (82%), the tumor was controlled without further treatment 19 - 87 months (median, 33 months) after SRT. Hypopituitarism was an adverse event after SRT in two of the 18 patients who achieved tumor control. Four patients (18%) had enlarged cysts after SRT. Postoperative pituitary function was significantly more likely to deteriorate in cases of extensive detachment from the ventricular wall, and retreatment was significantly more common in cases with hypothalamic extension. CONCLUSION: Although limited to some cases, ECPR combined with SRT is a less invasive and useful therapeutic option for suprasellar cystic craniopharyngiomas. However, its long-term prognosis requires further evaluation.


Craniopharyngioma , Neuroendoscopy , Pituitary Neoplasms , Radiosurgery , Humans , Craniopharyngioma/surgery , Craniopharyngioma/radiotherapy , Male , Female , Pituitary Neoplasms/surgery , Pituitary Neoplasms/radiotherapy , Adult , Middle Aged , Radiosurgery/methods , Radiosurgery/adverse effects , Neuroendoscopy/methods , Retrospective Studies , Treatment Outcome , Young Adult , Adolescent , Child , Cysts/surgery , Aged , Combined Modality Therapy/methods
4.
Eur J Pharm Biopharm ; 199: 114300, 2024 Jun.
Article En | MEDLINE | ID: mdl-38697488

Triple-negative breast cancer (TNBC) is considered one of the most incurable malignancies due to its clinical characteristics, including high invasiveness, high metastatic potential, proneness to relapse, and poor prognosis. Therefore, it remains a critical unmet medical need. On the other hand, poor delivery efficiency continues to reduce the efficacy of anti-cancer therapeutics developed against solid tumours using various strategies, such as genetically engineered oncolytic vectors used as nanocarriers. The study was designed to evaluate the anti-tumour efficacy of a novel combinatorial therapy based on oncolytic adenovirus AdV5/3-D24-ICOSL-CD40L with an anti-PD-1 (pembrolizumab) and paclitaxel (PTX). Here, we first tested the antineoplastic effect in two-dimensional (2D) and three-dimensional (3D) breast cancer models in MDA-MB-231, MDA-MB-468 and MCF-7 cells. Then, to further evaluate the efficacy of combinatorial therapy, including immunological aspects, we established a three-dimensional (3D) co-culture model based on MDA-MB-231 cells with peripheral blood mononuclear cells (PBMCs) to create an integrated system that more closely mimics the complexity of the tumour microenvironment and interacts with the immune system. Treatment with OV as a priming agent, followed by pembrolizumab and then paclitaxel, was the most effective in reducing the tumour volume in TNBC co-cultured spheroids. Further, T-cell phenotyping analyses revealed significantly increased infiltration of CD8+, CD4+ T and Tregs cells. Moreover, the observed anti-tumour effects positively correlated with the level of CD4+ T cell infiltrates, suggesting the development of anti-cancer immunity. Our study demonstrated that combining different immunotherapeutic agents (virus, pembrolizumab) with PTX reduced the tumour volume of the TNBC co-cultured spheroids compared to relevant controls. Importantly, sequential administration of the investigational agents (priming with the vector) further enhanced the anti-cancer efficacy in 3D culture over other groups tested. Taken together, these results support further evaluation of the virus in combination with anti-PD-1 and PTX for the treatment of triple-negative breast cancer patients. Importantly, further studies with in vivo models should be conducted to better understand the translational aspects of tested therapy.


Adenoviridae , Antibodies, Monoclonal, Humanized , Oncolytic Virotherapy , Paclitaxel , Programmed Cell Death 1 Receptor , Triple Negative Breast Neoplasms , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Humans , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/immunology , Female , Adenoviridae/genetics , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/administration & dosage , Oncolytic Virotherapy/methods , Cell Line, Tumor , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Oncolytic Viruses , MCF-7 Cells , Combined Modality Therapy/methods , Tumor Microenvironment/drug effects , Animals , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/administration & dosage
5.
Front Immunol ; 15: 1358478, 2024.
Article En | MEDLINE | ID: mdl-38698840

Introduction: Cancer combination treatments involving immunotherapies with targeted radiation therapy are at the forefront of treating cancers. However, dosing and scheduling of these therapies pose a challenge. Mathematical models provide a unique way of optimizing these therapies. Methods: Using a preclinical model of multiple myeloma as an example, we demonstrate the capability of a mathematical model to combine these therapies to achieve maximum response, defined as delay in tumor growth. Data from mice studies with targeted radionuclide therapy (TRT) and chimeric antigen receptor (CAR)-T cell monotherapies and combinations with different intervals between them was used to calibrate mathematical model parameters. The dependence of progression-free survival (PFS), overall survival (OS), and the time to minimum tumor burden on dosing and scheduling was evaluated. Different dosing and scheduling schemes were evaluated to maximize the PFS and optimize timings of TRT and CAR-T cell therapies. Results: Therapy intervals that were too close or too far apart are shown to be detrimental to the therapeutic efficacy, as TRT too close to CAR-T cell therapy results in radiation related CAR-T cell killing while the therapies being too far apart result in tumor regrowth, negatively impacting tumor control and survival. We show that splitting a dose of TRT or CAR-T cells when administered in combination is advantageous only if the first therapy delivered can produce a significant benefit as a monotherapy. Discussion: Mathematical models are crucial tools for optimizing the delivery of cancer combination therapy regimens with application along the lines of achieving cure, maximizing survival or minimizing toxicity.


Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Animals , Immunotherapy, Adoptive/methods , Mice , Combined Modality Therapy/methods , Receptors, Chimeric Antigen/immunology , Humans , Multiple Myeloma/therapy , Multiple Myeloma/immunology , Multiple Myeloma/radiotherapy , Models, Theoretical , Cell Line, Tumor , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/radiotherapy , Radioisotopes/therapeutic use , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
6.
Int J Mol Sci ; 25(9)2024 May 03.
Article En | MEDLINE | ID: mdl-38732225

Oncolytic viruses (OVs) are characterised by their preference for infecting and replicating in tumour cells either naturally or after genetic modification, resulting in oncolysis. Furthermore, OVs can elicit both local and systemic anticancer immune responses while specifically infecting and lysing tumour cells. These characteristics render them a promising therapeutic approach for paediatric brain tumours (PBTs). PBTs are frequently marked by a cold tumour immune microenvironment (TIME), which suppresses immunotherapies. Recent preclinical and clinical studies have demonstrated the capability of OVs to induce a proinflammatory immune response, thereby modifying the TIME. In-depth insights into the effect of OVs on different cell types in the TIME may therefore provide a compelling basis for using OVs in combination with other immunotherapy modalities. However, certain limitations persist in our understanding of oncolytic viruses' ability to regulate the TIME to enhance anti-tumour activity. These limitations primarily stem from the translational limitations of model systems, the difficulties associated with tracking reliable markers of efficacy throughout the course of treatment and the role of pre-existing viral immunity. In this review, we describe the different alterations observed in the TIME in PBTs due to OV treatment, combination therapies of OVs with different immunotherapies and the hurdles limiting the development of effective OV therapies while suggesting future directions based on existing evidence.


Brain Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Tumor Microenvironment , Humans , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Oncolytic Virotherapy/methods , Tumor Microenvironment/immunology , Oncolytic Viruses/physiology , Oncolytic Viruses/genetics , Child , Immunotherapy/methods , Combined Modality Therapy/methods , Animals
7.
Int J Nanomedicine ; 19: 4533-4568, 2024.
Article En | MEDLINE | ID: mdl-38799699

Until now, there has been a lack of effective strategies for cancer treatment. Immunotherapy has high potential in treating several cancers but its efficacy is limited as a monotherapy. Chemoimmunotherapy (CIT) holds promise to be widely used in cancer treatment. Therefore, identifying their involvement and potential synergy in CIT approaches is decisive. Nano-based drug delivery systems (NDDSs) are ideal delivery systems because they can simultaneously target immune cells and cancer cells, promoting drug accumulation, and reducing the toxicity of the drug. In this review, we first introduce five current immunotherapies, including immune checkpoint blocking (ICB), adoptive cell transfer therapy (ACT), cancer vaccines, oncolytic virus therapy (OVT) and cytokine therapy. Subsequently, the immunomodulatory effects of chemotherapy by inducing immunogenic cell death (ICD), promoting tumor killer cell infiltration, down-regulating immunosuppressive cells, and inhibiting immune checkpoints have been described. Finally, the NDDSs-mediated collaborative drug delivery systems have been introduced in detail, and the development of NDDSs-mediated CIT nanoparticles has been prospected.


Immunotherapy , Neoplasms , Humans , Immunotherapy/methods , Neoplasms/therapy , Neoplasms/immunology , Animals , Nanoparticles/chemistry , Cancer Vaccines/administration & dosage , Oncolytic Virotherapy/methods , Nanoparticle Drug Delivery System/chemistry , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/administration & dosage , Drug Delivery Systems/methods , Combined Modality Therapy/methods
8.
Cochrane Database Syst Rev ; 5: CD013042, 2024 May 28.
Article En | MEDLINE | ID: mdl-38802121

BACKGROUND: Manual therapy and prescribed exercises are often provided together or separately in contemporary clinical practice to treat people with lateral elbow pain. OBJECTIVES: To assess the benefits and harms of manual therapy, prescribed exercises or both for adults with lateral elbow pain. SEARCH METHODS: We searched the databases CENTRAL, MEDLINE and Embase, and trial registries until 31 January 2024, unrestricted by language or date of publication. SELECTION CRITERIA: We included randomised or quasi-randomised trials. Participants were adults with lateral elbow pain. Interventions were manual therapy, prescribed exercises or both. Primary comparators were placebo or minimal or no intervention. We also included comparisons of manual therapy and prescribed exercises with either intervention alone, with or without glucocorticoid injection. Exclusions were trials testing a single application of an intervention or comparison of different types of manual therapy or prescribed exercises. DATA COLLECTION AND ANALYSIS: Two review authors independently selected studies for inclusion, extracted trial characteristics and numerical data, and assessed study risk of bias and certainty of evidence using GRADE. The main comparisons were manual therapy, prescribed exercises or both compared with placebo treatment, and with minimal or no intervention. Major outcomes were pain, disability, heath-related quality of life, participant-reported treatment success, participant withdrawals, adverse events and serious adverse events. The primary endpoint was end of intervention for pain, disability, health-related quality of life and participant-reported treatment success and final time point for adverse events and withdrawals. MAIN RESULTS: Twenty-three trials (1612 participants) met our inclusion criteria (mean age ranged from 38 to 52 years, 47% female, 70% dominant arm affected). One trial (23 participants) compared manual therapy to placebo manual therapy, 12 trials (1124 participants) compared manual therapy, prescribed exercises or both to minimal or no intervention, six trials (228 participants) compared manual therapy and exercise to exercise alone, one trial (60 participants) compared the addition of manual therapy to prescribed exercises and glucocorticoid injection, and four trials (177 participants) assessed the addition of manual therapy, prescribed exercises or both to glucocorticoid injection. Twenty-one trials without placebo control were susceptible to performance and detection bias as participants were not blinded to the intervention. Other biases included selection (nine trials, 39%, including two quasi-randomised), attrition (eight trials, 35%) and selective reporting (15 trials, 65%) biases. We report the results of the main comparisons. Manual therapy versus placebo manual therapy Low-certainty evidence, based upon a single trial (23 participants) and downgraded due to indirectness and imprecision, indicates manual therapy may reduce pain and elbow disability at the end of two to three weeks of treatment. Mean pain at the end of treatment was 4.1 points with placebo (0 to 10 scale) and 2.0 points with manual therapy, MD -2.1 points (95% CI -4.2 to -0.1). Mean disability was 40 points with placebo (0 to 100 scale) and 15 points with manual therapy, MD -25 points (95% CI -43 to -7). There was no follow-up beyond the end of treatment to show if these effects were sustained, and no other major outcomes were reported. Manual therapy, prescribed exercises or both versus minimal intervention Low-certainty evidence indicates manual therapy, prescribed exercises or both may slightly reduce pain and disability at the end of treatment, but the effects were not sustained, and there may be little to no improvement in health-related quality of life or number of participants reporting treatment success. We downgraded the evidence due to increased risk of performance bias and detection bias across all the trials, and indirectness due to the multimodal nature of the interventions included in the trials. At four weeks to three months, mean pain was 5.10 points with minimal treatment and manual therapy, prescribed exercises or both reduced pain by a MD of -0.53 points (95% CI -0.92 to -0.14, I2 = 43%; 12 trials, 1023 participants). At four weeks to three months, mean disability was 63.8 points with minimal or no treatment and manual therapy, prescribed exercises or both reduced disability by a MD of -5.00 points (95% CI -9.22 to -0.77, I2 = 63%; 10 trials, 732 participants). At four weeks to three months, mean quality of life was 73.04 points with minimal treatment on a 0 to 100 scale and prescribed exercises reduced quality of life by a MD of -5.58 points (95% CI -10.29 to -0.99; 2 trials, 113 participants). Treatment success was reported by 42% of participants with minimal or no treatment and 57.1% of participants with manual therapy, prescribed exercises or both, RR 1.36 (95% CI 0.96 to 1.93, I2 = 73%; 6 trials, 770 participants). We are uncertain if manual therapy, prescribed exercises or both results in more withdrawals or adverse events. There were 83/566 participant withdrawals (147 per 1000) from the minimal or no intervention group, and 77/581 (126 per 1000) from the manual therapy, prescribed exercises or both groups, RR 0.86 (95% CI 0.66 to 1.12, I2 = 0%; 12 trials). Adverse events were mild and transient and included pain, bruising and gastrointestinal events, and no serious adverse events were reported. Adverse events were reported by 19/224 (85 per 1000) in the minimal treatment group and 70/233 (313 per 1000) in the manual therapy, prescribed exercises or both groups, RR 3.69 (95% CI 0.98 to 13.97, I2 = 72%; 6 trials). AUTHORS' CONCLUSIONS: Low-certainty evidence from a single trial in people with lateral elbow pain indicates that, compared with placebo, manual therapy may provide a clinically worthwhile benefit in terms of pain and disability at the end of treatment, although the 95% confidence interval also includes both an important improvement and no improvement, and the longer-term outcomes are unknown. Low-certainty evidence from 12 trials indicates that manual therapy and exercise may slightly reduce pain and disability at the end of treatment, but this may not be clinically worthwhile and these benefits are not sustained. While pain after treatment was an adverse event from manual therapy, the number of events was too small to be certain.


Bias , Exercise Therapy , Glucocorticoids , Musculoskeletal Manipulations , Randomized Controlled Trials as Topic , Tennis Elbow , Humans , Musculoskeletal Manipulations/methods , Exercise Therapy/methods , Glucocorticoids/therapeutic use , Tennis Elbow/therapy , Adult , Quality of Life , Middle Aged , Combined Modality Therapy/methods , Female , Injections, Intra-Articular
9.
Int J Mol Sci ; 25(8)2024 Apr 09.
Article En | MEDLINE | ID: mdl-38673726

Bone tumors, particularly osteosarcoma, are prevalent among children and adolescents. This ailment has emerged as the second most frequent cause of cancer-related mortality in adolescents. Conventional treatment methods comprise extensive surgical resection, radiotherapy, and chemotherapy. Consequently, the management of bone tumors and bone regeneration poses significant clinical challenges. Photothermal tumor therapy has attracted considerable attention owing to its minimal invasiveness and high selectivity. However, key challenges have limited its widespread clinical use. Enhancing the tumor specificity of photosensitizers through targeting or localized activation holds potential for better outcomes with fewer adverse effects. Combinations with chemotherapies or immunotherapies also present avenues for improvement. In this review, we provide an overview of the most recent strategies aimed at overcoming the limitations of photothermal therapy (PTT), along with current research directions in the context of bone tumors, including (1) target strategies, (2) photothermal therapy combined with multiple therapies (immunotherapies, chemotherapies, and chemodynamic therapies, magnetic, and photodynamic therapies), and (3) bifunctional scaffolds for photothermal therapy and bone regeneration. We delve into the pros and cons of these combination methods and explore current research focal points. Lastly, we address the challenges and prospects of photothermal combination therapy.


Bone Neoplasms , Infrared Rays , Photothermal Therapy , Humans , Bone Neoplasms/therapy , Photothermal Therapy/methods , Infrared Rays/therapeutic use , Animals , Photosensitizing Agents/therapeutic use , Osteosarcoma/therapy , Osteosarcoma/pathology , Combined Modality Therapy/methods , Immunotherapy/methods , Photochemotherapy/methods , Bone Regeneration
10.
J Cancer Res Ther ; 20(2): 540-546, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38687923

PURPOSE: To retrospectively study the therapeutic effect and safety performance of the combination strategies of the computed tomography (CT)-guided microwave ablation (MWA) and percutaneous vertebroplasty (PVP) as a treatment for painful non-small cell lung cancer (NSCLC) with spinal metastases. MATERIALS AND METHODS: A retrospective review included 71 patients with 109 vertebral metastases who underwent microwave ablation combined with percutaneous vertebroplasty by the image-guided and real-time temperature monitoring. Treatment efficacy was determined by comparing visual analog scale (VAS) scores, daily morphine equivalent opioid consumption, and Oswestry Disability Index (ODI) scores before treatment and during the follow-up period. RESULTS: Technical success was achieved in all patients. The mean pre-procedure VAS score and morphine doses were 6.6 ± 1.8 (4-10) and 137.2 ± 38.7 (40-200) mg, respectively. The mean VAS scores and daily morphine doses at 24 h and 1, 4, 12, and 24 weeks postoperatively were 3.3 ± 1.9 and 73.5 ± 39.4 mg; 2.2 ± 1.5 and 40.2 ± 29.8 mg; 1.7 ± 1.2 and 31.3 ± 23.6 mg; 1.4 ± 1.1 and 27.3 ± 21.4 mg; and 1.3 ± 1.1 and 24.8 ± 21.0 mg, respectively (all P < 0.001). ODI scores significantly decreased (P < 0.05). Minor cement leakage occurred in 51 cases (46.8%), with one patient having a grade 3 neural injury. No local tumor progression was observed by follow-up imaging. CONCLUSIONS: MWA combined with PVP can significantly relieve pain and improve patients' quality of life, which implied this is an effective treatment option for painful NSCLC with spinal metastases. Additionally, its efficacy should be further verified through the mid- and long-term studies.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Microwaves , Spinal Neoplasms , Vertebroplasty , Humans , Male , Female , Vertebroplasty/methods , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/surgery , Carcinoma, Non-Small-Cell Lung/secondary , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Middle Aged , Microwaves/therapeutic use , Spinal Neoplasms/secondary , Spinal Neoplasms/therapy , Spinal Neoplasms/complications , Aged , Retrospective Studies , Treatment Outcome , Combined Modality Therapy/methods , Pain Measurement , Tomography, X-Ray Computed , Adult , Cancer Pain/etiology , Cancer Pain/therapy , Cancer Pain/diagnosis , Aged, 80 and over , Pain Management/methods , Follow-Up Studies
11.
Drug Resist Updat ; 74: 101082, 2024 May.
Article En | MEDLINE | ID: mdl-38569225

Molecular targeted drugs and chimeric antigen receptor (CAR) T cell therapy represent specific biological treatments that have significantly improved the efficacy of treating hematologic malignancies. However, they face challenges such as drug resistance and recurrence after treatment. Combining molecular targeted drugs and CAR-T cells could regulate immunity, improve tumor microenvironment (TME), promote cell apoptosis, and enhance sensitivity to tumor cell killing. This approach might provide a dual coordinated attack on cancer cells, effectively eliminating minimal residual disease and overcoming therapy resistance. Moreover, molecular targeted drugs can directly or indirectly enhance the anti-tumor effect of CAR-T cells by inducing tumor target antigen expression, reversing CAR-T cell exhaustion, and reducing CAR-T cell associated toxic side effects. Therefore, combining molecular targeted drugs with CAR-T cells is a promising and novel tactic for treating hematologic malignancies. In this review article, we focus on analyzing the mechanism of therapy resistance and its reversal of CAR-T cell therapy resistance, as well as the synergistic mechanism, safety, and future challenges in CAR-T cell therapy in combination with molecular targeted drugs. We aim to explore the benefits of this combination therapy for patients with hematologic malignancies and provide a rationale for subsequent clinical studies.


Hematologic Neoplasms , Immunotherapy, Adoptive , Molecular Targeted Therapy , Tumor Microenvironment , Humans , Hematologic Neoplasms/therapy , Hematologic Neoplasms/immunology , Hematologic Neoplasms/drug therapy , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/trends , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Molecular Targeted Therapy/methods , Drug Resistance, Neoplasm/drug effects , Combined Modality Therapy/methods , Receptors, Chimeric Antigen/immunology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Animals
12.
Curr Treat Options Oncol ; 25(5): 628-643, 2024 May.
Article En | MEDLINE | ID: mdl-38649630

OPINION STATEMENT: Immunotherapy for glioblastoma (GBM) remains an intensive area of investigation. Given the seismic impact of cancer immunotherapy across a range of malignancies, there is optimism that harnessing the power of immunity will influence GBM as well. However, despite several phase 3 studies, there are still no FDA-approved immunotherapies for GBM. Importantly, the field has learned a great deal from the randomized studies to date. Today, we are continuing to better understand the disease-specific features of the microenvironment in GBM-as well as the exploitable antigenic characteristic of the tumor cells themselves-that are informing the next generation of immune-based therapeutic strategies. The coming phase of next-generation immunotherapies is thus poised to bring us closer to treatments that will improve the lives of patients with GBM.


Brain Neoplasms , Immunotherapy , Tumor Microenvironment , Humans , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Immunotherapy/methods , Tumor Microenvironment/immunology , Glioblastoma/therapy , Glioblastoma/immunology , Combined Modality Therapy/methods , Treatment Outcome , Disease Management , Clinical Trials as Topic
13.
Biomed Pharmacother ; 174: 116532, 2024 May.
Article En | MEDLINE | ID: mdl-38574625

Chimeric antigen receptor T (CAR-T) cell therapy, a groundbreaking immunotherapy. However, it faces formidable challenges in treating solid tumors, grappling with issues like poor trafficking, limited penetration, and insufficient persistence within the tumor microenvironment (TME). CAR-T cells are engineered to express receptors that target specific cancer antigens, enhancing their ability to recognize and eliminate cancer cells. This review paper explores the intricate interplay between CAR-T therapy and radiotherapy (RT), investigating their synergistic potential. Radiotherapy, a standard cancer treatment, involves using high doses of radiation to target and damage cancer cells, disrupting their ability to grow and divide. We highlight that RT modulates the TME, augments antigen presentation, and promotes immune cell infiltration, bolstering CAR-T cell-mediated tumor eradication. Molecular insights shed light on RT-induced alterations in tumor stroma, T cell recruitment promotion, and induction of immunogenic cell death. Noteworthy, strategies, such as combining hypofractionated radiotherapy with myeloid-derived suppressor cell blockade, underscore innovative approaches to enhance CAR-T cell therapy in solid tumors. Bridging indications for RT and CAR-T cells in hematological malignancies are discussed, emphasizing scenarios where RT strategically enhances CAR-T cell efficacy. The paper critically evaluates the RT as a bridge compared to traditional chemotherapy, highlighting timing and dosage considerations crucial for optimizing CAR-T therapy outcomes. In summary, the paper provides valuable insights into the intricate molecular mechanisms activated by RT and innovative strategies to improve CAR-T cell therapy, fostering a deeper understanding of their combined potential in cancer treatment.


Immunotherapy, Adoptive , Neoplasms , Tumor Microenvironment , Humans , Neoplasms/radiotherapy , Neoplasms/immunology , Neoplasms/pathology , Immunotherapy, Adoptive/methods , Animals , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Combined Modality Therapy/methods , Radiotherapy/methods
14.
Cancer Lett ; 591: 216858, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38621460

Radiotherapy (RT) remodels the tumor immune microenvironment (TIME) and modulates the immune response to indirectly destroy tumor cells, in addition to directly killing tumor cells. RT combined with immunotherapy may significantly enhance the efficacy of RT in colorectal cancer by modulating the microenvironment. However, the molecular mechanisms by which RT acts as an immunomodulator to modulate the immune microenvironment remain unclear. Further, the optimal modalities of RT combined with immunotherapy for the treatment of colorectal cancer, such as the time point of combining RT and immunization, the fractionation pattern and dosage of radiotherapy, and other methods to improve the efficacy, are also being explored parallelly. To address these aspects, in this review, we summarized the mechanisms by which RT modulates TIME and concluded the progress of RT combined with immunization in preclinical and clinical trials. Finally, we discussed heavy ion radiation therapy and the efficacy of prediction markers and other immune combination therapies. Overall, combining RT with immunotherapy to enhance antitumor effects will have a significant clinical implication and will help to facilitate individualized treatment modalities.


Colorectal Neoplasms , Immunotherapy , Tumor Microenvironment , Humans , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Colorectal Neoplasms/radiotherapy , Colorectal Neoplasms/pathology , Immunotherapy/methods , Tumor Microenvironment/immunology , Combined Modality Therapy/methods , Animals , Heavy Ion Radiotherapy/methods
15.
Curr Treat Options Oncol ; 25(5): 605-616, 2024 May.
Article En | MEDLINE | ID: mdl-38573430

OPINION STATEMENT: The treatment of oligometastatic genitourinary cancers is a rapidly advancing field with ablative radiotherapy as one of the critical treatment components. The oligometastatic disease state, which can be defined as 1-5 metastatic sites with a controlled primary, represents a distinct clinical state where comprehensive ablative local therapies may provide improved outcomes. Enhanced imaging has increased the number of patients identified with oligometastatic disease. Evidence for improved outcomes with metastasis-directed therapy (MDT) in oligometastatic genitourinary cancers is increasing, and previously published outcome data continues to mature with an increasing body of prospective data to inform the role of MDT in histology-specific settings or in the context of systemic therapy. In select patients, MDT can offer benefits beyond improved local control and allow for time off of systemic therapy, prolonged time until next therapy, or even the hope of cure. However, treatment decisions for locally ablative therapy must be balanced with consideration towards safety. There are exciting advances in technologies to target and adapt treatment in real-time which have expanded options for safer delivery and dose escalation to metastatic targets near critical organs at risk. The role of systemic therapies in conjunction with MDT and incorporation of tumor genetic information to further refine prognostication and treatment decision-making in the oligometastatic setting is actively being investigated. These developments highlight the evolving field of treatment of oligometastatic disease. Future prospective studies combining MDT with enhanced imaging and integrating MDT with evolving systemic therapies will enable the optimal selection of patients most likely to benefit from this "all-or-none" approach and reveal settings in which a combination of therapies could result in synergistic outcomes.


Neoplasm Metastasis , Urogenital Neoplasms , Humans , Urogenital Neoplasms/therapy , Urogenital Neoplasms/diagnosis , Urogenital Neoplasms/pathology , Disease Management , Combined Modality Therapy/methods , Treatment Outcome , Clinical Decision-Making
16.
Theranostics ; 14(6): 2573-2588, 2024.
Article En | MEDLINE | ID: mdl-38646638

Background: Hypofractionated radiotherapy (hRT) can induce a T cell-mediated abscopal effect on non-irradiated tumor lesions, especially in combination with immune checkpoint blockade (ICB). However, clinically, this effect is still rare, and ICB-mediated adverse events are common. Lenalidomide (lena) is an anti-angiogenic and immunomodulatory drug used in the treatment of hematologic malignancies. We here investigated in solid tumor models whether lena can enhance the abscopal effect in double combination with hRT. Methods: In two syngeneic bilateral tumor models (B16-CD133 melanoma and MC38 colon carcinoma), the primary tumor was treated with hRT. Lena was given daily for 3 weeks. Besides tumor size and survival, the dependence of the antitumor effects on CD8+ cells, type-I IFN signaling, and T cell costimulation was determined with depleting or blocking antibodies. Tumor-specific CD8+ T cells were quantified, and their differentiation and effector status were characterized by multicolor flow cytometry using MHC-I tetramers and various antibodies. In addition, dendritic cell (DC)-mediated tumor antigen cross-presentation in vitro and directly ex vivo and the composition of tumor-associated vascular endothelial cells were investigated. Results: In both tumor models, the hRT/lena double combination induced a significant abscopal effect. Control of the non-irradiated secondary tumor and survival were considerably better than with the respective monotherapies. The abscopal effect was strongly dependent on CD8+ cells and associated with an increase in tumor-specific CD8+ T cells in the non-irradiated tumor and its draining lymph nodes. Additionally, we found more tumor-specific T cells with a stem-like (TCF1+ TIM3- PD1+) and a transitory (TCF1- TIM3+ CD101- PD1+) exhausted phenotype and more expressing effector molecules such as GzmB, IFNγ, and TNFα. Moreover, in the non-irradiated tumor, hRT/lena treatment also increased DCs cross-presenting a tumor model antigen. Blocking type-I IFN signaling, which is essential for cross-presentation, completely abrogated the abscopal effect. A gene expression analysis of bone marrow-derived DCs revealed that lena augmented the expression of IFN response genes and genes associated with differentiation, maturation (including CD70, CD83, and CD86), migration to lymph nodes, and T cell activation. Flow cytometry confirmed an increase in CD70+ CD83+ CD86+ DCs in both irradiated and abscopal tumors. Moreover, the hRT/lena-induced abscopal effect was diminished when these costimulatory molecules were blocked simultaneously using antibodies. In line with the enhanced infiltration by DCs and tumor-specific CD8+ T cells, including more stem-like cells, hRT/lena also increased tumor-associated high endothelial cells (TA-HECs) in the non-irradiated tumor. Conclusions: We demonstrate that lena can augment the hRT-induced abscopal effect in mouse solid tumor models in a CD8 T cell- and IFN-I-dependent manner, correlating with enhanced anti-tumor CD8 T cell immunity, DC cross-presentation, and TA-HEC numbers. Our findings may be helpful for the planning of clinical trials in (oligo)metastatic patients.


CD8-Positive T-Lymphocytes , Disease Models, Animal , Lenalidomide , Radiation Dose Hypofractionation , Animals , Lenalidomide/pharmacology , Lenalidomide/therapeutic use , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Mice, Inbred C57BL , Dendritic Cells/immunology , Dendritic Cells/drug effects , Cell Line, Tumor , Combined Modality Therapy/methods , Female , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/radiotherapy , Melanoma, Experimental/therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/radiotherapy , Colonic Neoplasms/drug therapy , Colonic Neoplasms/therapy
17.
BMC Musculoskelet Disord ; 25(1): 328, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38658984

OBJECTIVE: To evaluate the clinical efficacy and imaging outcomes of percutaneous endoscopic lumbar discectomy (PELD) combined with platelet-rich plasma (PRP) for the treatment of lumbar disc herniation (LDH). METHODS: A total of 155 patients with LDH between January 2020 and June 2022 were retrospective analyzed, of which 75 underwent PELD with PRP and 80 underwent PELD only. Clinical functional scores and imaging data were compared. Clinical functional scores included visual analog scale of leg pain (VAS-LP) and back pain (VAS-BP), Japanese Orthopedic Association score (JOA), Oswestry Disability Index (ODI) and modified MacNab criteria. Imaging data included disc height index (DHI), spinal cross-sectional area (SCSA), disc protrusion size (DPZ), and ratio value of disc grey scales (RVG). RESULTS: Both groups showed clinical improvement, and VAS-LP, VAS-BP, JOA and ODI were significantly improved in the PRP group compared with the control group at 3, 6 and 12 months postoperatively (P < 0.05). At the last follow-up, the differences in SCSA, DPZ and RVG between the two groups were statistically significant (P < 0.05), with the PRP group being superior to the control group. The excellent and good rates of the modified Macnab criteria in the PRP group and control group were 93.3% and 90%, respectively, with no statistically significant difference (P > 0.05). No serious complications occurred during the follow-up period. CONCLUSION: PELD combined with PRP is a safe and effective method for treating patients with LDH. PRP injection was beneficial for delaying disc degeneration and promoting disc remodeling.


Diskectomy, Percutaneous , Endoscopy , Intervertebral Disc Displacement , Lumbar Vertebrae , Platelet-Rich Plasma , Humans , Intervertebral Disc Displacement/surgery , Intervertebral Disc Displacement/diagnostic imaging , Male , Female , Lumbar Vertebrae/surgery , Lumbar Vertebrae/diagnostic imaging , Diskectomy, Percutaneous/methods , Retrospective Studies , Adult , Middle Aged , Treatment Outcome , Endoscopy/methods , Pain Measurement , Combined Modality Therapy/methods , Magnetic Resonance Imaging
18.
J Cosmet Dermatol ; 23(6): 2015-2021, 2024 Jun.
Article En | MEDLINE | ID: mdl-38426374

BACKGROUND: Acne vulgaris is a common inflammatory disease associated with various sequelae after skin lesion remission. Acne erythema has been considered simple erythema or a vascular lesion; however, because the understanding of this disease has improved, acne erythema is currently considered an early scar with erythematous components. AIMS: This study evaluated the efficacy of using both a 595-nm pulsed dye laser (PDL) and 1565-nm nonablative fractional laser (NAFL) for the treatment of erythematous scars caused by acne. METHODS: Ninety patients with acne scars were equally randomized to two groups. Group A (n = 45) received treatment with the NAFL. Group B (n = 45) received treatment with the PDL and NAFL. Each patient underwent one treatment session and 4 weeks of follow-up. RESULTS: Qualitative (χ2 = 12.415; p < 0.05) and quantitative (t = 2.675; p < 0.05) scores of Groups A and B were determined using a global scarring grading system and exhibited statistically significant differences. The quantitative score of Group A was higher than that of Group B (6.67 ± 3.46 vs. 4.98 ± 2.44). The erythema areas of the groups differed significantly after treatment, with Group B exhibiting more notable score improvements (5.00 [3.10, 7.10] vs. 2.80 [1.65, 4.60]; Z = 3.072; p < 0.05). The erythema regression rate of Group B (88.9%) was significantly higher than that of Group A (66.7%) (χ2 = 20.295; p < 0.001). Adverse events, including redness and swelling (86.6%), scabbing (78.8%), and purpura (36.6%), occurred within 7 days for 86.6% of patients. CONCLUSIONS: The combined use of the PDL and NAFL is safe and effective for erythematous acne scars.


Acne Vulgaris , Cicatrix , Erythema , Lasers, Dye , Humans , Lasers, Dye/therapeutic use , Lasers, Dye/adverse effects , Acne Vulgaris/complications , Acne Vulgaris/radiotherapy , Cicatrix/etiology , Cicatrix/therapy , Cicatrix/diagnosis , Cicatrix/radiotherapy , Female , Male , Erythema/etiology , Adult , Young Adult , Treatment Outcome , Low-Level Light Therapy/adverse effects , Low-Level Light Therapy/methods , Low-Level Light Therapy/instrumentation , Lasers, Solid-State/therapeutic use , Lasers, Solid-State/adverse effects , Combined Modality Therapy/methods , Combined Modality Therapy/adverse effects , Severity of Illness Index , Adolescent
19.
J Cosmet Dermatol ; 23(6): 1992-2000, 2024 Jun.
Article En | MEDLINE | ID: mdl-38429946

BACKGROUND: Acne scar is an inflammatory condition, which commonly occurs in patients with acne vulgaris, especially in adults. Mesogels have been reported effective in improving atrophic acne scars. AIMS: We investigated the efficacy of adding Profhilo (a hyaluronic acid-based filler) to subcision as a new treatment method. METHODS: Twelve patients aged 18-45 years with atrophic acne scars on both sides of the face participated in this single-blinded, split-face, randomized controlled trial. Each side of the face was randomly assigned to one of the treatment methods, including subcision alone and subcision + Profhilo. Patients in the Profhilo arm received mesogel (1 cc) in addition to the subcision procedure. Both methods were carried out two times at 1-month intervals. Assessments were done based on the sonographic depth of scars, and two blinded observers examined photographs at baseline and 3 months after the final session and the results were reported based on an exclusively made formula as the total score. The Global Improvement Scale and Visual Analogue Scale (VAS) (for patient satisfaction) were also used. RESULTS: The VAS score of patient satisfaction was statistically significant in the Profhilo arm, with a mean improvement of 528.08 and 219.06 in the subcision arm (p = 0.02). No significant difference was seen in total acne scar reduction comparing the two methods (29.74 in the Profhilo arm and 22.27 in the subcision arm, p = 0.56). Sonographic depth reduction was also non-significant, with a mean of 29.21 in the Profhilo arm and 28.53 in the subcision arm (p = 0.4). The mean global improvement was reported as four in both arms, and no statistical significance was observed (p = 0.89). The best response to treatment belonged to the rolling subtype in both methods (p = 0.029 for the Profhilo arm and p = 0.001 for the subcision arm). CONCLUSION: Despite no significant difference between the methods, Profhilo is more effective due to a higher satisfaction rate and better physiologic effects.


Acne Vulgaris , Cicatrix , Dermal Fillers , Hyaluronic Acid , Patient Satisfaction , Humans , Acne Vulgaris/complications , Single-Blind Method , Adult , Cicatrix/etiology , Cicatrix/therapy , Female , Hyaluronic Acid/administration & dosage , Young Adult , Male , Treatment Outcome , Dermal Fillers/administration & dosage , Dermal Fillers/adverse effects , Adolescent , Middle Aged , Combined Modality Therapy/methods , Face , Cosmetic Techniques/adverse effects , Cosmetic Techniques/instrumentation
20.
J Cosmet Dermatol ; 23(6): 2049-2057, 2024 Jun.
Article En | MEDLINE | ID: mdl-38456556

BACKGROUND: There is no standardized and effective treatment modality for Riehl's melanosis. AIMS: To compare the efficacy and safety of oral tranexamic acid (TXA) combined with intense pulsed light (IPL) versus TXA alone in the treatment of refractory Riehl's melanosis. METHODS: A prospective study of 28 subjects with refractory Riehl's melanosis and Fitzpatrick Skin Types III or IV was conducted. All subjects received oral TXA 500 mg daily and 11 of them were treated in combination with monthly IPL therapy for 6 months. The primary outcome measure was mean melanin index (MI), erythema index (EI) and acquired dermal macular hyperpigmentation area and severity index (DPASI). The Physician Global Assessment (PGA) and patient satisfaction scale were documented. RESULTS: After treatment, DPASI, mean MI, and EI were significantly reduced in both groups. The group treated with combination therapy showed better improvement according to MI (p = 0.0032) and DPASI (p = 0.00468). PGA and patient satisfaction scale showed superior efficacy in the combination group. No significant difference was observed in treatment-related side effects. CONCLUSION: The combination of oral TXA and IPL proves to be a safe and satisfactory treatment strategy for refractory Riehl's melanosis.


Antifibrinolytic Agents , Intense Pulsed Light Therapy , Melanosis , Patient Satisfaction , Tranexamic Acid , Humans , Tranexamic Acid/administration & dosage , Tranexamic Acid/adverse effects , Melanosis/therapy , Melanosis/drug therapy , Melanosis/diagnosis , Prospective Studies , Female , Male , Middle Aged , Adult , Administration, Oral , Treatment Outcome , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Intense Pulsed Light Therapy/adverse effects , Intense Pulsed Light Therapy/methods , Antifibrinolytic Agents/administration & dosage , Antifibrinolytic Agents/adverse effects , Severity of Illness Index
...