Network


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

Hotspot


Dive into the research topics where Scott Knowles is active.

Publication


Featured researches published by Scott Knowles.


Journal of Clinical Oncology | 2012

Advances in Immuno–Positron Emission Tomography: Antibodies for Molecular Imaging in Oncology

Scott Knowles; Anna M. Wu

Identification of cancer cell-surface biomarkers and advances in antibody engineering have led to a sharp increase in the development of therapeutic antibodies. These same advances have led to a new generation of radiolabeled antibodies and antibody fragments that can be used as cancer-specific imaging agents, allowing quantitative imaging of cell-surface protein expression in vivo. Immuno-positron emission tomography (immunoPET) imaging with intact antibodies has shown success clinically in diagnosing and staging cancer. Engineered antibody fragments, such as diabodies, minibodies, and single-chain Fv (scFv) -Fc, have been successfully employed for immunoPET imaging of cancer cell-surface biomarkers in preclinical models and are poised to bring same-day imaging into clinical development. ImmunoPET can potentially provide a noninvasive approach for obtaining target-specific information useful for titrating doses for radioimmunotherapy, for patient risk stratification and selection of targeted therapies, for evaluating response to therapy, and for predicting adverse effects, thus contributing to the ongoing development of personalized cancer treatment.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Engineered antibody fragments for immuno-PET imaging of endogenous CD8+ T cells in vivo

Richard Tavaré; Melissa N. McCracken; Kirstin A. Zettlitz; Scott Knowles; Felix B. Salazar; Tove Olafsen; Owen N. Witte; Anna M. Wu

Significance Anti-CD8 immuno-PET imaging agents provide the potential to monitor the localization, migration, and expansion of CD8-expressing cells noninvasively in vivo. Shown here is the successful generation of functional anti-CD8 imaging agents based on engineered antibodies for use in a variety of preclinical disease and immunotherapeutic models. The noninvasive detection and quantification of CD8+ T cells in vivo are important for both the detection and staging of CD8+ lymphomas and for the monitoring of successful cancer immunotherapies, such as adoptive cell transfer and antibody-based immunotherapeutics. Here, antibody fragments are constructed to target murine CD8 to obtain rapid, high-contrast immuno-positron emission tomography (immuno-PET) images for the detection of CD8 expression in vivo. The variable regions of two anti-murine CD8-depleting antibodies (clones 2.43 and YTS169.4.2.1) were sequenced and reformatted into minibody (Mb) fragments (scFv-CH3). After production and purification, the Mbs retained their antigen specificity and bound primary CD8+ T cells from the thymus, spleen, lymph nodes, and peripheral blood. Importantly, engineering of the parental antibodies into Mbs abolished the ability to deplete CD8+ T cells in vivo. The Mbs were subsequently conjugated to S-2-(4-isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid for 64Cu radiolabeling. The radiotracers were injected i.v. into antigen-positive, antigen-negative, immunodeficient, antigen-blocked, and antigen-depleted mice to evaluate specificity of uptake in lymphoid tissues by immuno-PET imaging and ex vivo biodistribution. Both 64Cu-radiolabeled Mbs produced high-contrast immuno-PET images 4 h postinjection and showed specific uptake in the spleen and lymph nodes of antigen-positive mice.


The Journal of Nuclear Medicine | 2014

Quantitative ImmunoPET of Prostate Cancer Xenografts with 89Zr- and 124I-Labeled Anti-PSCA A11 Minibody

Scott Knowles; Kirstin A. Zettlitz; Richard Tavaré; Matthew M. Rochefort; Felix B. Salazar; David Stout; Paul J. Yazaki; Robert E. Reiter; Anna M. Wu

Prostate stem cell antigen (PSCA) is expressed on the cell surface in 83%–100% of local prostate cancers and 87%–100% of prostate cancer bone metastases. In this study, we sought to develop immunoPET agents using 124I- and 89Zr-labeled anti-PSCA A11 minibodies (scFv-CH3 dimer, 80 kDa) and evaluate their use for quantitative immunoPET imaging of prostate cancer. Methods: A11 anti-PSCA minibody was alternatively labeled with 124I- or 89Zr-desferrioxamine and injected into mice bearing either matched 22Rv1 and 22Rv1×PSCA or LAPC-9 xenografts. Small-animal PET data were obtained and quantitated with and without recovery coefficient–based partial-volume correction, and the results were compared with ex vivo biodistribution. Results: Rapid and specific localization to PSCA-positive tumors and high-contrast imaging were observed with both 124I- and 89Zr-labeled A11 anti-PSCA minibody. However, the differences in tumor uptake and background uptake of the radiotracers resulted in different levels of imaging contrast. The nonresidualizing 124I-labeled minibody had lower tumor uptake (3.62 ± 1.18 percentage injected dose per gram [%ID/g] 22Rv1×PSCA, 3.63 ± 0.59 %ID/g LAPC-9) than the residualizing 89Zr-labeled minibody (7.87 ± 0.52 %ID/g 22Rv1×PSCA, 9.33 ± 0.87 %ID/g LAPC-9, P < 0.0001 for each), but the 124I-labeled minibody achieved higher imaging contrast because of lower nonspecific uptake and better tumor–to–soft-tissue ratios (22Rv1×PSCA:22Rv1 positive-to-negative tumor, 13.31 ± 5.59 124I-A11 and 4.87 ± 0.52 89Zr-A11, P = 0.02). Partial-volume correction was found to greatly improve the correspondence between small-animal PET and ex vivo quantification of tumor uptake for immunoPET imaging with both radionuclides. Conclusion: Both 124I- and 89Zr-labeled A11 anti-PSCA minibody showed high-contrast imaging of PSCA expression in vivo. However, the 124I-labeled A11 minibody was found to be the superior imaging agent because of lower nonspecific uptake and higher tumor–to–soft-tissue contrast. Partial-volume correction was found to be essential for robust quantification of immunoPET imaging with both 124I- and 89Zr-labeled A11 minibody.


Protein Engineering Design & Selection | 2010

Tuning the serum persistence of human serum albumin domain III:diabody fusion proteins.

Tove Olafsen; Felix B. Salazar; Lawrence E. Williams; Scott Knowles; Anna M. Wu

The long circulation persistence of human serum albumin (HSA) is enabled by its domain III (DIII) interaction with the neonatal Fc receptor (FcRn). A protein scaffold based on HSA DIII was designed. To modify the serum half life of the scaffold, residues H535, H510, and H464 were individually mutated to alanine. HSA DIII wild type (WT) and variants were fused to the anti-carcinoembryonic antigen (CEA) T84.66 diabody (Db), radiolabeled with (124)I and injected into xenografted athymic mice for serial PET/CT imaging. All proteins targeted the CEA-positive tumor. The mean residence times (MRT) of the proteins, calculated by quantifying blood activity from the PET images, were: Db-DIII WT (56.7 h), H535A (25 h), H510A (20 h), H464A (17 h), compared with Db (2.9 h). Biodistribution confirmed the order of blood clearance from slow to fast: Db-DIII WT > H535A > H510A > H464A > Db with 4.0, 2.0, 1.8, 1.6 and 0.08 %ID/g of remaining blood activity at 51 h, respectively. This study demonstrates that attenuating the DIII-FcRn interaction provides a way of controlling the pharmacokinetics of the entire Db-DIII fusion protein without compromising tumor targeting. H464 appears to be most crucial for FcRn binding (greatest reduction in MRT), followed by H510 and H535. By mutating the DIII scaffold, we can dial serum kinetics for imaging or therapy applications.


Biomacromolecules | 2013

Endocytosis and intracellular trafficking properties of transferrin-conjugated block copolypeptide vesicles.

Uh-Joo Choe; April R. Rodriguez; Brian S. Lee; Scott Knowles; Anna M. Wu; Timothy J. Deming; Daniel T. Kamei

Block polypeptides are an emerging class of materials that have the potential to be used in many biomedical applications, including the field of drug delivery. We have previously developed a negatively charged block copolypeptide, poly(L-glutamate)60-b-poly(L-leucine)20 (E60L20), which forms spherical vesicles in aqueous solution. Since these vesicles are negatively charged, they are minimally toxic toward cells. However, the negative charge also inhibits these vesicles from effectively being internalized by cells, which can be problematic as many therapeutics have intracellular targets. To overcome this limitation of the E60L20 vesicles, transferrin (Tf) was conjugated onto the vesicle surface, since the receptor for Tf is overexpressed on cancer cells. The enhanced uptake of the Tf-conjugated vesicle was verified through confocal microscopy. Furthermore, endocytosis and immunostaining experiments confirmed that the Tf conjugated on the vesicle surface plays a critical role in the internalization and subsequent intracellular trafficking behavior of the vesicles.


Clinical Cancer Research | 2014

Applications of ImmunoPET: Using 124I-Anti-PSCA A11 Minibody for Imaging Disease Progression and Response to Therapy in Mouse Xenograft Models of Prostate Cancer

Scott Knowles; Richard Tavaré; Kirstin A. Zettlitz; Matthew M. Rochefort; Felix B. Salazar; Ziyue Karen Jiang; Robert E. Reiter; Anna M. Wu

Purpose: Prostate stem cell antigen (PSCA) is highly expressed in local prostate cancers and prostate cancer bone metastases and its expression correlates with androgen receptor activation and a poor prognosis. In this study, we investigate the potential clinical applications of immunoPET with the anti-PSCA A11 minibody, an antibody fragment optimized for use as an imaging agent. We compare A11 minibody immunoPET to 18F-Fluoride PET bone scans for detecting prostate cancer bone tumors and evaluate the ability of the A11 minibody to image tumor response to androgen deprivation. Experimental Design: Osteoblastic, PSCA-expressing, LAPC-9 intratibial xenografts were imaged with serial 124I-anti-PSCA A11 minibody immunoPET and 18F-Fluoride bone scans. Mice bearing LAPC-9 subcutaneous xenografts were treated with either vehicle or MDV-3100 and imaged with A11 minibody immunoPET/CT scans pre- and posttreatment. Ex vivo flow cytometry measured the change in PSCA expression in response to androgen deprivation. Results: A11 minibody demonstrated improved sensitivity and specificity over 18F-Fluoride bone scans for detecting LAPC-9 intratibial xenografts at all time points. LAPC-9 subcutaneous xenografts showed downregulation of PSCA when treated with MDV-3100 which A11 minibody immunoPET was able to detect in vivo. Conclusions: A11 minibody immunoPET has the potential to improve the sensitivity and specificity of clinical prostate cancer metastasis detection over bone scans, which are the current clinical standard-of-care. A11 minibody immunoPET additionally has the potential to image the activity of the androgen signaling axis in vivo which may help evaluate the clinical response to androgen deprivation and the development of castration resistance. Clin Cancer Res; 20(24); 6367–78. ©2014 AACR.


Clinical Cancer Research | 2016

Fluorescent Image–Guided Surgery with an Anti-Prostate Stem Cell Antigen (PSCA) Diabody Enables Targeted Resection of Mouse Prostate Cancer Xenografts in Real Time

Geoffrey A. Sonn; Andrew S. Behesnilian; Ziyue Karen Jiang; Kirstin A. Zettlitz; Eric J. Lepin; Laurent A. Bentolila; Scott Knowles; Daniel J P Lawrence; Anna M. Wu; Robert E. Reiter

Purpose: The inability to visualize cancer during prostatectomy contributes to positive margins, cancer recurrence, and surgical side effects. A molecularly targeted fluorescent probe offers the potential for real-time intraoperative imaging. The goal of this study was to develop a probe for image-guided prostate cancer surgery. Experimental Design: An antibody fragment (cys-diabody, cDb) against prostate stem cell antigen (PSCA) was conjugated to a far-red fluorophore, Cy5. The integrity and binding of the probe to PSCA was confirmed by gel electrophoresis, size exclusion, and flow cytometry, respectively. Subcutaneous models of PSCA-expressing xenografts were used to assess the biodistribution and in vivo kinetics, whereas an invasive intramuscular model was utilized to explore the performance of Cy5-cDb–mediated fluorescence guidance in representative surgical scenarios. Finally, a prospective, randomized study comparing surgical resection with and without fluorescent guidance was performed to determine whether this probe could reduce the incidence of positive margins. Results: Cy5-cDb demonstrated excellent purity, stability, and specific binding to PSCA. In vivo imaging showed maximal signal-to-background ratios at 6 hours. In mice carrying PSCA+ and negative (−) dual xenografts, the mean fluorescence ratio of PSCA+/− tumors was 4.4:1. In surgical resection experiments, residual tumors <1 mm that were missed on white light surgery were identified and resected using fluorescence guidance, which reduced the incidence of positive surgical margins (0/8) compared with white light surgery alone (7/7). Conclusions: Fluorescently labeled cDb enables real-time in vivo imaging of prostate cancer xenografts in mice, and facilitates more complete tumor removal than conventional white light surgery alone. Clin Cancer Res; 22(6); 1403–12. ©2015 AACR. See related commentary by van Leeuwen and van der Poel, p. 1304


The Journal of Nuclear Medicine | 2014

Improved Modeling of In Vivo Kinetics of Slowly Diffusing Radiotracers for Tumor Imaging

Moses Q. Wilks; Scott Knowles; Anna M. Wu; Sung-Cheng Huang

Large-molecule tracers, such as labeled antibodies, have shown success in immuno-PET for imaging of specific cell surface biomarkers. However, previous work has shown that localization of such tracers shows high levels of heterogeneity in target tissues, due to both the slow diffusion and the high affinity of these compounds. In this work, we investigate the effects of subvoxel spatial heterogeneity on measured time–activity curves in PET imaging and the effects of ignoring diffusion limitation on parameter estimates from kinetic modeling. Methods: Partial differential equations (PDE) were built to model a radially symmetric reaction-diffusion equation describing the activity of immuno-PET tracers. The effects of slower diffusion on measured time–activity curves and parameter estimates were measured in silico, and a modified Levenberg–Marquardt algorithm with Bayesian priors was developed to accurately estimate parameters from diffusion-limited data. This algorithm was applied to immuno-PET data of mice implanted with prostate stem cell antigen–overexpressing tumors and injected with 124I-labeled A11 anti–prostate stem cell antigen minibody. Results: Slow diffusion of tracers in linear binding models resulted in heterogeneous localization in silico but no measurable differences in time–activity curves. For more realistic saturable binding models, measured time–activity curves were strongly dependent on diffusion rates of the tracers. Fitting diffusion-limited data with regular compartmental models led to parameter estimate bias in an excess of 1,000% of true values, while the new model and fitting protocol could accurately measure kinetics in silico. In vivo imaging data were also fit well by the new PDE model, with estimates of the dissociation constant (Kd) and receptor density close to in vitro measurements and with order of magnitude differences from a regular compartmental model ignoring tracer diffusion limitation. Conclusion: Heterogeneous localization of large, high-affinity compounds can lead to large differences in measured time–activity curves in immuno-PET imaging, and ignoring diffusion limitations can lead to large errors in kinetic parameter estimates. Modeling of these systems with PDE models with Bayesian priors is necessary for quantitative in vivo measurements of kinetics of slow-diffusion tracers.


Journal of Laboratory Automation | 2017

Engineering A11 Minibody-Conjugated, Polypeptide-Based Gold Nanoshells for Prostate Stem Cell Antigen (PSCA)-Targeted Photothermal Therapy.

Kristine M. Mayle; Kathryn R. Dern; Vincent K. Wong; Kevin Y. Chen; Shijun Sung; Ke Ding; April R. Rodriguez; Scott Knowles; Zachary D. Taylor; Z. Hong Zhou; Warren S. Grundfest; Anna M. Wu; Timothy J. Deming; Daniel T. Kamei

Currently, there is no curative treatment for advanced metastatic prostate cancer, and options, such as chemotherapy, are often nonspecific, harming healthy cells and resulting in severe side effects. Attaching targeting ligands to agents used in anticancer therapies has been shown to improve efficacy and reduce nonspecific toxicity. Furthermore, the use of triggered therapies can enable spatial and temporal control over the treatment. Here, we combined an engineered prostate cancer–specific targeting ligand, the A11 minibody, with a novel photothermal therapy agent, polypeptide-based gold nanoshells, which generate heat in response to near-infrared light. We show that the A11 minibody strongly binds to the prostate stem cell antigen that is overexpressed on the surface of metastatic prostate cancer cells. Compared to nonconjugated gold nanoshells, our A11 minibody-conjugated gold nanoshell exhibited significant laser-induced, localized killing of prostate cancer cells in vitro. In addition, we improved upon a comprehensive heat transfer mathematical model that was previously developed by our laboratory. By relaxing some of the assumptions of our earlier model, we were able to generate more accurate predictions for this particular study. Our experimental and theoretical results demonstrate the potential of our novel minibody-conjugated gold nanoshells for metastatic prostate cancer therapy.


Cancer Research | 2011

Abstract 4283: Evaluation of PSCA-specific minibody for in vivo imaging

Chau P. Tran; Scott Knowles; Tove Olafsen; Eric J. Lepin; Anna M. Wu; Robert E. Reiter

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL Introduction: Molecular targeted therapy of advanced prostate cancer requires the ability to classify tumors at the molecular level without invasive tissue sampling. Noninvasive molecular imaging can yield information on tumor localization, phenotype, and response to therapy. Previously we have generated humanized antibody to prostate stem cell antigen (PSCA), a cell surface marker highly elevated in prostate cancers, and used the antibody to detect tumors by PET imaging. To improve affinity, minibody variants were obtained by yeast display, and one clone, A11, demonstrated high imaging contrast in PSCA positive xenograft tumors. However, xenograft tumors are limited as model of natural disease progression, and cannot be used to assess background uptake of antibody in normal tissues, given that the humanized antibody does not recognize the murine (m)PSCA. This study aims to evaluate the biodistribution of A11 minibody in a mouse model expressing human (h)PSCA. Such model is important for assessing minibody performance prior to further clinical development. Methods: hPSCA knock-in (KI) mouse was generated by replacing exon 1 of mPSCA with hPSCA cDNA thereby driving hPSCA expression under mPSCA promoter. hPSCA-KI line was evaluated for hPSCA expression in major organs by RT-PCR and immunohistochemistry. For imaging study, groups of 8 weeks old hPSCA-KI and wild-type mice were injected with radiolabeled (I-124) minibody A11 and imaged by microPET at 4 and 20 hrs post injection. Mice were then sacrificed and major organs collected, weighed, and counted to determine biodistribution of A11. Results: By immunohistochemistry, hPSCA is expressed in prostate, bladder and stomach of heterozygous hPSCA-KI mice as expected. Other organs showed much lower or no expression of hPSCA, as confirmed by RT-PCR. MicroPET imaging showed specific uptake of I-124 labeled A11 in the prostate, bladder and stomach of hPSCA-KI mice. A11 biodistribution (% injected dose/gram of tissue) was significantly higher in hPSCA-KI mice compared to wild-type mice: prostate (0.83 ± 0.13 vs. 0.49 ± 0.04), bladder (3.21 ± 0.42 vs. 1.66 ± 0.25) and stomach (2.00 ± 0.22 vs. 0.79 ± 0.05). There was no uptake difference between groups for other major organs including pancreas, lung, kidney, spleen and liver. Conclusions: The minibody uptake in hPSCA-KI mice was specific to normal site of expression (prostate, bladder and stomach). While the % uptake was significantly higher than wild-type controls, the actual levels were modest and should not necessarily interfere with tumor imaging. This part of the study remains to be investigated. PSCA has been shown to be elevated in the PTEN conditional knockout prostate cancer mouse model. Thus, hPSCA-KI;PTEN-knockout mice are being generated and will be used to evaluate the PSCA-specific minibody as a tumor imaging tool. PSCA minibody imaging may be useful as a surrogate to diagnose and monitor response to targeted therapy of this type of prostate cancers. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4283. doi:10.1158/1538-7445.AM2011-4283

Collaboration


Dive into the Scott Knowles's collaboration.

Top Co-Authors

Avatar

Anna M. Wu

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tove Olafsen

University of California

View shared research outputs
Top Co-Authors

Avatar

Eric J. Lepin

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Stout

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Moses Q. Wilks

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge