Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 241
Filter
1.
Int J Hyperthermia ; 40(1): 2164625, 2023.
Article in English | MEDLINE | ID: mdl-36966808

ABSTRACT

BACKGROUND: It has been demonstrated that cryoablation (Cryo) causes specific T-cell immune responses in the body; however, it is not sufficient to prevent tumor recurrence and metastasis. In this report, we evaluated changes in the tumor immune microenvironment (TIME) in distant tumor tissues after Cryo and investigated the immunosuppressive mechanisms that limit the efficacy of Cryo. METHODS: Bilateral mammary tumor models were established in mice, and we first observed the dynamic changes in immune cells and cytokines at different time points after Cryo. Then, we confirmed that the upregulation of PD-1 and PD-L1 signaling in the contralateral tumor tissue was closely related to the immunosuppressive state in the TIME at the later stage after Cryo. Finally, we also evaluated the synergistic antitumor effects of Cryo combined with PD-1 monoclonal antibody (mAb) in the treatment of breast cancer (BC) mouse. RESULTS: We found that Cryo can stimulate the body's immune response, but it also induces immunosuppression. The elevated PD-1/PD-L1 expression in distant tumor tissues at the later stage after Cryo was closely related to the immunosuppressive state in the TIME but also created the conditions for Cryo combined with PD-1 mAb for BC mouse treatment. Cryo + PD-1 mAb could improve the immunosuppressive state of tumors and enhance the Cryo-induced immune response, thus exerting a synergistic antitumor effect. CONCLUSIONS: The PD-1/PD-L1 axis plays an important role in suppressing Cryo-induced antitumor immune responses. This study provides a theoretical basis for Cryo combined with PD-1 mAb therapy in clinical BC patients.


Subject(s)
Antibodies, Monoclonal , Cryosurgery , Mammary Neoplasms, Experimental , Animals , Mice , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/pharmacology , Cell Line, Tumor , Disease Models, Animal , Neoplasm Recurrence, Local/drug therapy , Programmed Cell Death 1 Receptor/metabolism , Tumor Microenvironment , Mammary Neoplasms, Animal , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/surgery
2.
Proc Natl Acad Sci U S A ; 119(15): e2123111119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35380898

ABSTRACT

In vivo fluorescence/luminescence imaging in the near-infrared-IIb (NIR-IIb, 1,500 to 1,700 nm) window under <1,000 nm excitation can afford subcentimeter imaging depth without any tissue autofluorescence, promising high-precision intraoperative navigation in the clinic. Here, we developed a compact imager for concurrent visible photographic and NIR-II (1,000 to 3,000 nm) fluorescence imaging for preclinical image-guided surgery. Biocompatible erbium-based rare-earth nanoparticles (ErNPs) with bright down-conversion luminescence in the NIR-IIb window were conjugated to TRC105 antibody for molecular imaging of CD105 angiogenesis markers in 4T1 murine breast tumors. Under a ∼940 ± 38 nm light-emitting diode (LED) excitation, NIR-IIb imaging of 1,500- to 1,700-nm emission afforded noninvasive tumor­to­normal tissue (T/NT) signal ratios of ∼40 before surgery and an ultrahigh intraoperative tumor-to-muscle (T/M) ratio of ∼300, resolving tumor margin unambiguously without interfering background signal from surrounding healthy tissues. High-resolution imaging resolved small numbers of residual cancer cells during surgery, allowing thorough and nonexcessive tumor removal at the few-cell level. NIR-IIb molecular imaging afforded 10-times-higher and 100-times-higher T/NT and T/M ratios, respectively, than imaging with IRDye800CW-TRC105 in the ∼900- to 1,300-nm range. The vastly improved resolution of tumor margin and diminished background open a paradigm of molecular imaging-guided surgery.


Subject(s)
Erbium , Mammary Neoplasms, Experimental , Metal Nanoparticles , Optical Imaging , Spectroscopy, Near-Infrared , Surgery, Computer-Assisted , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Fluorescence , Fluorescent Dyes/chemistry , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/surgery , Mice , Neoplasm, Residual/diagnostic imaging , Optical Imaging/methods , Spectroscopy, Near-Infrared/methods , Surgery, Computer-Assisted/methods
3.
PLoS One ; 16(9): e0256076, 2021.
Article in English | MEDLINE | ID: mdl-34495961

ABSTRACT

Thermal therapies such as radiofrequency ablation (RFA) are gaining widespread clinical adoption in the local treatment of skeletal metastases. RFA has been shown to successfully destroy tumor cells, yet the impact of RFA on the quality of the surrounding bone has not been well characterized. RFA treatment was performed on femora of rats with bone metastases (osteolytic and osteoblastic) and healthy age matched rats. Histopathology, second harmonic generation imaging and backscatter electron imaging were used to characterize changes in the structure, organic and mineral components of the bone after RFA. RFA treatment was shown to be effective in targeting tumor cells and promoting subsequent new bone formation without impacting the surrounding bone negatively. Mineralization profiles of metastatic models were significantly improved post-RFA treatment with respect to mineral content and homogeneity, suggesting a positive impact of RFA treatment on the quality of cancer involved bone. Evaluating the impact of RFA on bone quality is important in directing the growth of this minimally invasive therapeutic approach with respect to fracture risk assessment, patient selection, and multimodal treatment planning.


Subject(s)
Bone Neoplasms/secondary , Bone Neoplasms/surgery , Calcification, Physiologic , Mammary Neoplasms, Experimental/surgery , Radiofrequency Ablation , Animals , Bone Neoplasms/metabolism , Disease Models, Animal , Female , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Rats , Rats, Nude , Tomography, X-Ray Computed , Treatment Outcome
4.
Anticancer Res ; 40(9): 4869-4874, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32878774

ABSTRACT

BACKGROUND/AIM: In the present study, we evaluated the efficacy of adjuvant administration of oral recombinant methioninase (o-rMETase) against recurrence and metastasis in a 4T1 murine breast-cancer syngeneic model. MATERIALS AND METHODS: 4T1 cells were orthotopically implanted into the 2nd mammary fat pad of BALB/c mice. The 4T1 orthotopic syngeneic models were randomized into 2 groups after primary tumor resection: untreated control and o-rMETase (100 units, oral, daily, 2 weeks). RESULTS: The frequency and extent of local recurrence were reduced by o-rMETase. The number of individual cancer cells and metastatic nodules on the lung surface was significantly lower in the o-rMETase-treated mice than the untreated control mice. CONCLUSION: Adjuvant o-rMETase inhibited local recurrence and lung metastasis after primary tumor resection.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Carbon-Sulfur Lyases/administration & dosage , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/drug therapy , Administration, Oral , Animals , Cell Line, Tumor , Chemotherapy, Adjuvant , Drug Administration Schedule , Female , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/surgery , Mice , Mice, Inbred BALB C , Neoplasm Recurrence, Local/prevention & control , Recombinant Proteins/administration & dosage , Xenograft Model Antitumor Assays
5.
Opt Lett ; 45(7): 2006-2009, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32236054

ABSTRACT

Laser ablation (LA) is a promising approach for minimally invasive cancer treatments. Its in vivo applicability is often impeded by the lack of efficient monitoring tools that can help to minimize collateral tissue damage and aid in determining the optimal treatment end-points. We have devised a new, to the best of our knowledge, hybrid LA approach combining simultaneous volumetric optoacoustic (OA) imaging to monitor the lesion progression accurately in real time and 3D. Time-lapse imaging of laser ablation of solid tumors was performed in a murine breast cancer model in vivo by irradiation of subcutaneous tumors with a 100 mJ short-pulsed (${\sim}{5}\;{\rm ns}$∼5ns) laser operating at 1064 nm and 100 Hz pulse repetition frequency. Local changes in the OA signal intensity ascribed to structural alterations in the tumor vasculature were clearly observed, while the OA volumetric projections recorded in vivo appeared to correlate with cross sections of the excised tumors.


Subject(s)
Laser Therapy , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/surgery , Photoacoustic Techniques/methods , Surgery, Computer-Assisted/methods , Animals , Female , Mice
6.
Clin Cancer Res ; 26(4): 910-921, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31757878

ABSTRACT

PURPOSE: Radiotherapy (RT) is frequently used for local control of solid tumors using equal dose per fraction. Recently, single high-dose radiation has been used for ablation of solid tumors. In this report, we provide a novel immunological basis for radiation dose fractionation consisting of a single high-dose radiotherapy, followed by postablation modulation (PAM) with four daily low-dose fractions (22 Gy + 0.5 Gy × 4) to reprogram the tumor microenvironment by diminishing immune suppression, enabling infiltration of effector cells and increasing efficacy of tumor control. EXPERIMENTAL DESIGN: Palpable 3LL and 4T1 tumors in C57Bl/6 and Balb/c mice were irradiated with the Small-Animal Radiation Research Platform irradiator, and tumor growth and survival were monitored. Immunomodulation of tumor and immune cells in vitro and in vivo characterization of tumor-infiltrating immune effector cells were performed by FACS. For systemic application of PAM-RT, whole-lung irradiation was administered in 4T1-bearing Balb/c mice. RESULTS: We report significant tumor growth delays and increased survival in 3LL tumor-bearing mice with PAM. Primary tumor PAM-RT increased infiltration of immune effector cells and decreased Treg in irradiated tumors and secondary lymphoid organs. In a model of murine metastatic breast cancer (4T1), we demonstrated that systemic PAM-RT to the whole lung, 12 days after primary tumor ablative radiotherapy, increased survival with suppression of pulmonary metastases. CONCLUSIONS: We provide a novel immunologic basis for radiation dose fractionation consisting of a single high dose of radiotherapy followed by daily low-dose PAM-RT fractionation to improve the immunogenic potential of ablative radiotherapy.


Subject(s)
Carcinoma, Lewis Lung/radiotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Mammary Neoplasms, Experimental/radiotherapy , Tumor Microenvironment/immunology , Ablation Techniques/methods , Animals , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/surgery , Cell Line, Tumor , Dose Fractionation, Radiation , Female , Immunomodulation/radiation effects , Lymphocytes, Tumor-Infiltrating/radiation effects , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/surgery , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Survival Rate , Tumor Microenvironment/radiation effects
7.
Chem Commun (Camb) ; 55(95): 14287-14290, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31712798

ABSTRACT

A novel bright near-infrared II (NIR-II, 1000-1700 nm) fluorescent probe with excellent water-solubility, superior photostability, and excellent in vitro and in vivo biocompatibility was facilely synthesized for in vivo biomedical imaging of xenograft breast tumor and chemically induced spontaneous breast carcinoma. To the best of our knowledge, it is the first time that the superior practical applications of this NIR-II probe in dimethylbenzanthracene (DMBA)-induced rat mammary carcinoma imaging and image-guided rat carcinoma surgery were demonstrated.


Subject(s)
Biocompatible Materials/chemistry , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/surgery , Fluorescent Dyes/chemistry , 9,10-Dimethyl-1,2-benzanthracene , Animals , Breast Neoplasms/chemically induced , Female , Humans , Infrared Rays , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/surgery , Mice , Mice, Inbred Strains , Rats , Rats, Sprague-Dawley
8.
PLoS One ; 14(7): e0207107, 2019.
Article in English | MEDLINE | ID: mdl-31269078

ABSTRACT

Breast cancer rates are rising in low- and middle-income countries (LMICs), yet there is a lack of accessible and cost-effective treatment. As a result, the cancer burden and death rates are highest in LMICs. In an effort to meet this need, our work presents the design and feasibility of a low-cost cryoablation system using widely-available carbon dioxide as the only consumable. This system uses an 8-gauge outer-diameter needle and Joule-Thomson expansion to percutaneously necrose tissue with cryoablation. Bench top experiments characterized temperature dynamics in ultrasound gel demonstrated that isotherms greater than 2 cm were formed. Further, this system was applied to mammary tumors in an in vivo rat model and necrosis was verified by histopathology. Finally, freezing capacity under a large heat load was assessed with an in vivo porcine study, where volumes of necrosis greater than 1.5 cm in diameter confirmed by histopathology were induced in a highly perfused liver after two 7-minute freeze cycles. These results demonstrate the feasibility of a carbon-dioxide based cryoablation system for improving solid tumor treatment options in resource-constrained environments.


Subject(s)
Carbon Dioxide/therapeutic use , Catheter Ablation , Cryosurgery , Mammary Neoplasms, Experimental/surgery , Animals , Catheter Ablation/instrumentation , Catheter Ablation/methods , Cryosurgery/instrumentation , Cryosurgery/methods , Female , Mammary Neoplasms, Experimental/pathology , Rats , Rats, Sprague-Dawley , Swine
9.
Sci Rep ; 9(1): 9204, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31235775

ABSTRACT

Angiogenesis is a key tumor microenvironment (TME) event underpinning tumor growth and metastasis. Nevertheless, the relatively poor performance of anti-angiogenic therapies in clinical trials compared to pre-clinical studies implies that classical subcutaneous xenograft models have limited predictive potential in this setting. To address this issue, we established orthotopic surgical resection models of breast cancer, which replicate the phenotype of clinical post-resection micro-metastasis. To demonstrate the power and precision of these models, we recapitulated the BETH adjuvant trial (NCT00625898) where the addition of bevacizumab (BVZ) to chemotherapy plus trastuzumab (Trast) failed to provide additional benefit. SCID mice were orthotopically implanted with bioluminescent Her2+ MDA-MB-231 or HCC1954 cells and tumors resected c.5 weeks later. Following resection, mice were treated with 10 mg/kg Trast +5 mg/kg paclitaxel (PAC) IP once weekly for 6 cycles +/- weekly BVZ (5 mg/kg IP). Metastasis was monitored by imaging. Using these models our data confirms that the addition of the anti-angiogenic antibody BVZ to adjuvant Trast + chemotherapy provides no additional benefit compared with Trast + chemotherapy alone. Previous studies using non-resection subcutaneously engrafted xenografts failed to predict this outcome. Our results provide compelling evidence for the utility of cell line xenograft resection models to predict clinical outcome for TME targeting agents.


Subject(s)
Bevacizumab/therapeutic use , Breast Neoplasms/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Paclitaxel/therapeutic use , Trastuzumab/therapeutic use , Animals , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/surgery , Cell Line, Tumor , Chemoradiotherapy, Adjuvant , Female , Humans , Mammary Neoplasms, Experimental/surgery , Mice , Mice, SCID , Transplantation, Heterologous , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
10.
J Healthc Eng ; 2018: 5806753, 2018.
Article in English | MEDLINE | ID: mdl-29854360

ABSTRACT

Microwave ablation (MWA) by using coaxial antennas is a promising alternative for breast cancer treatment. A double short distance slot coaxial antenna as a newly optimized applicator for minimally invasive treatment of breast cancer is proposed. To validate and to analyze the feasibility of using this method in clinical treatment, a computational model, phantom, and breast swine in vivo experimentation were carried out, by using four microwave powers (50 W, 30 W, 20 W, and 10 W). The finite element method (FEM) was used to develop the computational model. Phantom experimentation was carried out in breast phantom. The in vivo experimentation was carried out in a 90 kg swine sow. Tissue damage was estimated by comparing control and treated micrographs of the porcine mammary gland samples. The coaxial slot antenna was inserted in swine breast glands by using image-guided ultrasound. In all cases, modeling, in vivo and phantom experimentation, and ablation temperatures (above 60°C) were reached. The in vivo experiments suggest that this new MWA applicator could be successfully used to eliminate precise and small areas of tissue (around 20-30 mm2). By modulating the power and time applied, it may be possible to increase/decrease the ablation area.


Subject(s)
Breast Neoplasms/surgery , Catheter Ablation/instrumentation , Microwaves , Animals , Catheter Ablation/methods , Computer Simulation , Equipment Design , Feasibility Studies , Female , Finite Element Analysis , Humans , Mammary Neoplasms, Experimental/surgery , Minimally Invasive Surgical Procedures/instrumentation , Phantoms, Imaging , Swine , Temperature
11.
Br J Anaesth ; 121(1): 76-85, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29935598

ABSTRACT

BACKGROUND: Breast cancer accounts for 7% of female cancer deaths, usually attributable to metastasis. While surgery is a mainstay of treatment, perioperative interventions may influence risk of metastasis during breast tumour resection. Amide local anaesthetics influence cancer cell biology via numerous mechanisms in vitro, but in vivo data is lacking. We aimed to test the hypothesis that perioperative lidocaine reduces pulmonary metastasis after inhalation and i.v. anaesthesia in the 4T1 murine breast cancer model. METHODS: 4T1 Cancer cells were injected into the mammary fat-pad of immunocompetent BALB/c female mice. After 7 days, the resultant tumour was excised under either sevoflurane or ketamine/xylazine anaesthesia with or without perioperative i.v. lidocaine (1.5 mg kg-1 bolus followed by 25 min infusion 2 mg kg-1 h-1). Fourteen days post-surgery, posthumous lung and liver specimens were examined for metastasis. Pro-inflammatory and pro-metastatic cytokines were profiled in post-mortem serum from a small number of the mice. RESULTS: Primary tumour diameter was similar between groups. Lidocaine reduced lung metastatic colony count vs sevoflurane alone; median (inter-quartile range) 0 (0-2) compared with 22.5 (0-481), P=0.02 and reduced the proportion of animals with pulmonary metastasis (28.5% compared with 52.5%, P=0.04). In mice receiving ketamine-xylazine, lidocaine did not decrease the overall colony count: 60 (26-123) compared with 23.5 (0-225), P=0.43, but increased the proportion of animals with pulmonary metastasis (100% compared with 50%, P<0.01). Post-mortem serum analysis demonstrated reduced pro-inflammatory and angiogenic cytokine expression in animals without metastasis which received lidocaine with sevoflurane. CONCLUSIONS: In this 4T1 murine model of breast cancer, lidocaine decreased pulmonary metastasis when combined with sevoflurane anaesthesia, perhaps via anti-inflammatory and anti-angiogenic effects. It had no such effect in mice given ketamine anaesthesia.


Subject(s)
Adrenergic alpha-Agonists , Anesthetics, Dissociative , Anesthetics, Inhalation , Anesthetics, Local/pharmacology , Ketamine , Lidocaine/pharmacology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/surgery , Neoplasm Metastasis/prevention & control , Sevoflurane , Xylazine , Animals , Cell Line, Tumor , Cytokines/blood , Female , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control
12.
Acta Biomater ; 74: 302-311, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29729897

ABSTRACT

Locoregional recurrence of breast cancer after tumor resection represents several clinical challenges. Here, we demonstrate that co-delivery of chemotherapy and thermotherapeutic agents by a magnetic supramolecular hydrogel (MSH) following tumor resection prevents tumor recurrence in a breast cancer mouse model. The self-assembled MSH was designed through the partial inclusion complexation associated with the threading of α-CD on the copolymer moieties on the surface of the PEGylated iron oxide (Fe3O4) nanoparticles, which enables shear-thinning injection and controllable thermoreversible gel-sol transition. MSH was injected to the postoperative wound uniformly, which became mobile and perfect match with irregular cavity without blind angle due to the magnetocaloric gel-sol transition when exposed to alternating current magnetic field (ACMF). The magnetic nanoparticle-mediated induction heat during the gel-sol transition process caused the triggered release of dual-encapsulated chemotherapeutic drugs and provided an effect of thermally induced cell damage. The hierarchical structure of the MSH ensured that both hydrophobic and hydrophilic drugs can be loaded and consecutively delivered with different release curves. The hydrogel nanocomposite might provide a potential locally therapeutic approach for the precise treatment of locoregional recurrence of cancer. STATEMENT OF SIGNIFICANCE: Tumor recurrence after resection represents several clinical challenges. In this study, we prepared shear-thinning injectable magnetic supramolecular hydrogel (MSH) and demonstrated their therapeutic applications in preventing the post-operative recurrence of breast cancer with facile synthesis and minimally invasive implantation in vivo. MSH was injected to the postoperative wound uniformly, which become mobile and perfect match with irregular cavity without blind angle through magnetocaloric gel-sol transition when exposed to ACMF. The magnetic nanoparticles mediated induction heat during the gel-sol transition process caused the triggered release of dual-encapsulated chemotherapeutic drugs as well as thermally induced cell damage. This study demonstrates that MSH with the controlled administration of combined thermo-chemotherapy exhibit great superiority in terms of preventing post-operation cancer relapse.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Hydrogels , Magnetic Fields , Magnetite Nanoparticles , Mammary Neoplasms, Experimental , Neoplasm Recurrence, Local/prevention & control , Animals , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Female , Hydrogels/chemistry , Hydrogels/pharmacokinetics , Hydrogels/pharmacology , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/surgery , Mice , Mice, Inbred BALB C , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , RAW 264.7 Cells
13.
Vet Comp Oncol ; 16(1): 47-54, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28370855

ABSTRACT

BACKGROUND: The important goal of modern research in the field of surgical oncology is the quest for a tool that could improve the outcomes of tumour excision. AIMS: The aim of this study was to compare the usefulness of the CO2 laser with flexible hollow waveguide and scalpel in mammary tumour excision. MATERIALS & METHODS: A total of 112 female BALB/c mice with implanted orthotopically 4T1-luc2-tdTomato tumour cells were included in the research. Tumours were excised in 48 mice using the CO2 laser and in 48 through scalpel surgery. The control group consisted of 16 untreated mice. The evaluation of surgical outcome was obtained by in vivo bioluminescence and fluorescence imaging and post-mortem histopathological examination. RESULTS: There were no significant differences between recurrence rates, metastases and survival time in groups excised with the scalpel and CO2 laser. CONCLUSION: The CO2 laser has similar efficacy compared with conventional scalpel excision for local recurrence rates, incidence of distant metastases and survival time and can be safely applied in oncological surgery.


Subject(s)
Adenocarcinoma/veterinary , Lasers, Gas/therapeutic use , Mammary Neoplasms, Experimental/surgery , Adenocarcinoma/surgery , Animals , Female , Mammary Glands, Animal/surgery , Mice, Inbred BALB C , Neoplasm Recurrence, Local/veterinary , Neoplasm Transplantation/veterinary , Treatment Outcome
14.
Mol Pharm ; 15(3): 750-758, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29172524

ABSTRACT

Molecularly targeted optical contrast agents have the potential to enable surgeons to visualize specific molecular markers that can help improve surgical precision and thus outcomes. Fluorescently quenched substrates can be used to highlight tumor lesions by targeting proteases that are highly abundant in the tumor microenvironment. However, the majority of these and other molecularly targeted optical contrast agents are labeled with reporter dyes that are not ideally matched to the properties of clinical camera systems, which are typically optimized for detection of indocyanine-green (ICG). While a wide range of near-infrared (NIR) dyes are suitable for use with highly sensitive and highly tunable research-focused small animal imaging systems, most have not been evaluated for use with commonly used clinical imaging systems. Here we report the optimization of a small molecule fluorescently quenched protease substrate probe 6QC-ICG, which uses the indocyanine green (ICG) dye as its optical reporter. We evaluated dosing and kinetic parameters of this molecule in tumor-bearing mice and observed optimal tumor over background signals in as little as 90 min with a dose of 2.3 mg/kg. Importantly, the fluorescence intensity of the probe signal in tumors did not linearly scale with dose, suggesting the importance of detailed dosing studies. Furthermore, when imaged using the FDA approved da Vinci Si surgical system with Firefly detection, signals were significantly higher for the ICG probe compared to a corresponding probe containing a dye with similar quantum yield but with a slightly shifted excitation and emission profile. The increased signal intensity generated by the optimal dye and dose of the ICG labeled probe enabled detection of small, flat lesions that were less than 5 mm in diameter. Therefore, 6QC-ICG is a highly sensitive probe that performs optimally with clinical imaging systems and has great potential for applications in optical surgical navigation.


Subject(s)
Contrast Media/metabolism , Mammary Neoplasms, Experimental/diagnostic imaging , Neoplasms/diagnostic imaging , Peptide Hydrolases/metabolism , Video-Assisted Surgery/methods , Animals , Cell Line, Tumor/transplantation , Contrast Media/administration & dosage , Contrast Media/chemistry , Female , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Indocyanine Green/administration & dosage , Indocyanine Green/chemistry , Indocyanine Green/metabolism , Mammary Neoplasms, Experimental/surgery , Mice , Mice, Inbred BALB C , Molecular Imaging/methods , Neoplasms/pathology , Neoplasms/surgery
15.
Theranostics ; 7(19): 4722-4734, 2017.
Article in English | MEDLINE | ID: mdl-29187899

ABSTRACT

RATIONALE: Positive margin status due to incomplete removal of tumor tissue during breast conserving surgery (BCS) is a prevalent diagnosis usually requiring a second surgical procedure. These follow-up procedures increase the risk of morbidity and delay the use of adjuvant therapy; thus, significant efforts are underway to develop new intraoperative strategies for margin assessment to eliminate re-excision procedures. One strategy under development uses topical application of dual probe staining and a fluorescence imaging strategy termed dual probe difference specimen imaging (DDSI). DDSI uses a receptor-targeted fluorescent probe and an untargeted, spectrally-distinct fluorescent companion imaging agent topically applied to fresh resected specimens, where the fluorescence from each probe is imaged and a normalized difference image is computed to identify tumor-target distribution in the specimen margins. While previous reports suggested this approach is a promising new tool for surgical guidance, advancing the approach into the clinic requires methodical protocol optimization and further validation. METHODS: In the present study, we used breast cancer xenografts and receiver operator characteristic (ROC) curve analysis to evaluate a wide range of staining and imaging parameters, and completed a prospective validation study on multiple tumor phenotypes with different target expression. Imaging fluorophore-probe pair, concentration, and incubation times were systematically optimized using n=6 tissue specimen replicates per staining condition. Resulting tumor vs. normal adipose tissue diagnostic performance were reported and staining patterns were validated via receptor specific immunohistochemistry colocalization. Optimal staining conditions were tested in receptor positive and receptor negative cohorts to confirm specificity. RESULTS: The optimal staining conditions were found to be a one minute stain in a 200 nM probe solution (area under the curve (AUC) = 0.97), where the choice of fluorescent label combination did not significantly affect the diagnostic performance. Using an optimal threshold value determined from ROC curve analysis on a training data set, a prospective study on xenografts resulted in an AUC=0.95 for receptor positive tumors and an AUC = 0.50 for receptor negative (control) tumors, confirming the diagnostic performance of this novel imaging technique. CONCLUSIONS: DDSI provides a robust, molecularly specific imaging methodology for identifying tumor tissue over benign mammary adipose tissue. Using a dual probe imaging strategy, nonspecific accumulation of targeted probe was corrected for and tumor vs. normal tissue diagnostic potential was improved, circumventing difficulties with ex vivo tissue specimen staining and allowing for rapid clinical translation of this promising technology for tumor margin detection during BCS procedures.


Subject(s)
Biomarkers, Tumor/metabolism , Mammary Neoplasms, Experimental/surgery , Mastectomy, Segmental/methods , Staining and Labeling/methods , Surgery, Computer-Assisted/methods , Animals , Female , Fluorescent Dyes , Humans , MCF-7 Cells , Mastectomy, Segmental/standards , Mice , Mice, Nude , Sensitivity and Specificity , Staining and Labeling/standards , Surgery, Computer-Assisted/standards
16.
Int J Nanomedicine ; 12: 8337-8351, 2017.
Article in English | MEDLINE | ID: mdl-29200846

ABSTRACT

An intraoperative technique to accurately identify microscopic tumor residuals could decrease the risk of positive surgical margins. Several lines of evidence support the expression and immunotherapeutic effect of PD-1 in breast cancer. Here, we sought to develop a fluorescence-labeled PD-1 probe for in vivo breast tumor imaging and image-guided surgery. The efficacy of PD-1 monoclonal antibody (PD-1 mAb) as adjuvant immunotherapy after surgery was also assessed. PD-1-IRDye800CW was developed and examined for its application in tumor imaging and image-guided tumor resection in an immunocompetent 4T1 mouse tumor model. Fluorescence molecular imaging was performed to monitor probe biodistribution and intraoperative imaging. Bioluminescence imaging was performed to monitor tumor growth and evaluate postsurgical tumor residuals, recurrences, and metastases. The PD-1-IRDye800CW exhibited a specific signal at the tumor region compared with the IgG control. Furthermore, PD-1-IRDye800CW-guided surgery combined with PD-1 adjuvant immunotherapy inhibited tumor regrowth and microtumor metastases and thus improved survival rate. Our study demonstrates the feasibility of using PD-1-IRDye800CW for breast tumor imaging and image-guided tumor resection. Moreover, PD-1 mAb adjuvant immunotherapy reduces cancer recurrences and metastases emanating from tumor residuals.


Subject(s)
Fluorescent Dyes/chemistry , Mammary Neoplasms, Experimental/therapy , Monitoring, Intraoperative/methods , Programmed Cell Death 1 Receptor/chemistry , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/metabolism , Female , Fluorescent Dyes/pharmacokinetics , Fluorescent Dyes/therapeutic use , Immunotherapy/methods , Luminescent Measurements/methods , Mammary Neoplasms, Experimental/mortality , Mammary Neoplasms, Experimental/surgery , Mice, Inbred BALB C , Molecular Imaging/methods , Monitoring, Intraoperative/instrumentation , Optical Imaging/methods , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Surgery, Computer-Assisted/methods , Tissue Distribution
17.
Bull Exp Biol Med ; 164(2): 186-190, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29177878

ABSTRACT

The anterior mediastinal lymph nodes were analyzed morphometrically in rats with chemically provoked breast cancer. Rats with untreated breast cancer and animals receiving chemotherapy demonstrated decreased volumes of paracortical region and lymphoid nodules with the germinal centers accompanied by extended medullary thymic substance. Resection of largest focus of breast tumor improved the filtration barrier potential of anterior mediastinal lymph nodes, up-regulated the proliferative activity of lymphoid cells in T-cell zones, and down-regulated proliferation of plasmatic cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Lymph Nodes/pathology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/pathology , Mediastinum/pathology , Animals , Carcinogens/administration & dosage , Cell Proliferation/drug effects , Cyclophosphamide/pharmacology , Female , Fluorouracil/pharmacology , Injections, Subcutaneous , Lymph Nodes/drug effects , Lymph Nodes/surgery , Lymphatic Metastasis , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/surgery , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/surgery , Mediastinum/surgery , Methotrexate/pharmacology , Methylnitrosourea/administration & dosage , Plasma Cells/drug effects , Plasma Cells/pathology , Rats , Rats, Wistar , T-Lymphocytes/drug effects , T-Lymphocytes/pathology
18.
J Surg Res ; 219: 310-318, 2017 11.
Article in English | MEDLINE | ID: mdl-29078898

ABSTRACT

BACKGROUND: Despite massive expenditures in preclinical studies, many breast cancer agents show efficacy in murine models but fail in human trials. In humans, metastatic disease determines survival, but preclinical murine models only evaluate drug efficacy against the primary tumor. We hypothesized that evaluating efficacy against metastatic breast cancer would more efficiently predict efficacy in a murine model than evaluating the primary tumor alone. This study (1) critically evaluated a murine tumor removal model with metastatic tumor burden quantification for breast cancer preclinical trials and (2) validated the model with an agent that previously passed preclinical trials but failed human trials. MATERIALS AND METHODS: Tumorectomy and Halsted (radical) mastectomy procedures after inoculation of 4T1-luc2 cells were compared. The effect of AZD0530, an oral Src inhibitor that passed preclinical trials but failed human trials, was evaluated using an inoculation model with/without Halsted mastectomy. RESULTS: Significant amounts of residual disease were confirmed by bioluminescence (P = 0.003) and 100% developed local recurrence after tumorectomy versus 14% (P = 0.005) after Halsted mastectomy. Bioluminescence value at 15 min after luciferin injection highly correlated with peak except for 24 h after injection. AZD0530 significantly suppressed primary tumor burden compared with no treatment (P = 0.002); but not in lung metastases. In a Halsted mastectomy model, AZD0530 had no efficacy against lung metastases or difference in survival. CONCLUSIONS: We critically evaluated and established a murine mastectomy model to evaluate metastatic tumors. It provides a new model for preclinical drug development that mimics the human adjuvant setting.


Subject(s)
Adenocarcinoma/secondary , Antineoplastic Agents/therapeutic use , Benzodioxoles/therapeutic use , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/surgery , Mastectomy, Radical , Quinazolines/therapeutic use , Adenocarcinoma/drug therapy , Animals , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Luminescent Measurements , Lung/pathology , Lung Neoplasms/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Neoplasm Metastasis
19.
J Vis Exp ; (125)2017 07 29.
Article in English | MEDLINE | ID: mdl-28784976

ABSTRACT

A rate-limiting aspect of transgenic mouse models of mammary adenocarcinoma is that primary tumor burden in mammary tissue typically defines study end-points. Thus, studies focused on elucidating mechanisms of late-stage de novo metastasis are compromised, as are studies examining efficacy of anti-cancer therapies targeting mediators of metastasis in the adjuvant setting. Numerous murine mammary cancer models have been developed via targeted expression of dominant oncoproteins to mammary epithelial cells yielding models variably mimicking histopathologic and transcriptome-defined breast cancer subtypes common in women1. While much has been learned regarding the biology of mammary carcinogenesis with these models, their utility in identifying molecules regulating growth of late-stage metastasis are compromised as mice are typically euthanized at earlier time points due to significant primary tumor burden. Moreover, since a significant percentage of women diagnosed with breast cancer receive adjuvant therapy after surgical resection of primary tumors and prior to presence of detectable metastatic disease, preclinical models of de novo metastasis are urgently needed as platforms to evaluate new therapies aimed at targeting metastatic foci. To address these deficiencies, we developed a murine model of de novo mammary cancer metastasis, wherein primary mammary tumors are surgically resected, and metastatic foci subsequently develop over a 115 day post-surgical period. This long latency provides a tractable model to identify functionally significant regulators of metastatic progression in mice lacking primary tumor, as well as a model to evaluate preclinical therapeutic efficacy of agents aimed at blocking functionally significant molecules aiding metastatic tumor survival and growth.


Subject(s)
Mammary Neoplasms, Experimental/pathology , Surgical Procedures, Operative/methods , Animals , Disease Models, Animal , Female , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lymph Nodes/pathology , Lymphatic Metastasis , Mammary Neoplasms, Experimental/surgery , Mice , Mice, Transgenic , Neoplasm Recurrence, Local , Receptors, Virus/genetics , Video Recording
20.
Anticancer Res ; 37(6): 2965-2974, 2017 06.
Article in English | MEDLINE | ID: mdl-28551634

ABSTRACT

AIM: Effects of radiofrequency ablation (RFA) combining immunoadjuvant glycated chitosan (GC) on tumor control and potent cytokine responses were investigated in a syngeneic breast tumor model. MATERIALS AND METHODS: Murine 4T1 breast carcinoma cells harboring the luciferase reporter gene were used to evaluate the tumor growth rate and metastasis in vivo using bioluminescent imaging. Plasma of RFA/GC-treated tumor-bearing mice was collected for ex vivo cytotoxicity analysis and mouse chemokine array assays. RESULTS: Tumor growth and systemic metastasis were suppressed by combined RFA and GC when tumor size reached 300 mm3, not detected, however, when tumor size reached 800 mm3 The survival rate of mice bearing small tumors was also higher than that of large ones after RFA-GC treatment. Plasma extracted from RFA-GC-treated small tumor-bearing mice exhibited cytotoxicity on cultured 4T1 cells. Moreover, reduced tumor growth-related cytokines and increased antitumor-related cytokines were detected in the plasma collected. CONCLUSION: RFA combining GC could control tumor progression with induced potent antitumor cytokine responses.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Catheter Ablation , Chitosan/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/surgery , Animals , Cell Line, Tumor , Cell Survival , Chemokines/blood , Combined Modality Therapy , Female , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Plasma , Tumor Burden/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...