% \documentclass[twocolumn]{article} \documentclass{report} % TODO I want to keep figures in each subsection, which this doesn't do \usepackage[section]{placeins} \usepackage[top=1in,left=1.5in,right=1in,bottom=1in]{geometry} \usepackage{siunitx} \usepackage{multicol} \setlength{\columnsep}{1cm} \usepackage[acronym]{glossaries} \usepackage[T1]{fontenc} \usepackage{enumitem} \usepackage{titlesec} \usepackage{titlecaps} \usepackage{upgreek} \usepackage{graphicx} \usepackage{subcaption} \usepackage{nth} \usepackage[capitalize]{cleveref} \usepackage[version=4]{mhchem} \usepackage{pgfgantt} \usepackage{setspace} % TODO glossary can't apparently be used in section header (even thought it % would be nice) \doublespacing{} \titleformat{\chapter}[block]{\filcenter\bfseries\large} {\MakeUppercase{\chaptertitlename} \thechapter: }{0pt}{\uppercase} % \titleformat{\chapter}[block]{\filcenter\bfseries\large}{}{0pt}{\uppercase} \titleformat{\section}[block]{\bfseries\large}{}{0pt}{\titlecap} \titleformat{\subsection}[block]{\itshape\large}{}{0pt}{\titlecap} \titleformat{\subsubsection}[runin]{\bfseries\itshape\/}{}{0pt}{\titlecap} \setlist[description]{font=$\bullet$~\textbf\normalfont} %%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%% % acronyms for the lazy \renewcommand{\glossarysection}[2][]{} % remove glossary title \makeglossaries \newacronym{act}{ACT}{adoptive cell therapies} \newacronym{qc}{QC}{quality control} \newacronym{tcm}{T\textsubscript{cm}}{central memory T cell} \newacronym{tscm}{T\textsubscript{scm}}{stem-memory T cell} \newacronym{car}{CAR}{chimeric antigen receptor} \newacronym[longplural={monoclonal antibodies}]{mab}{mAb}{monoclonal antibody} \newacronym{ecm}{ECM}{extracellular matrix} \newacronym{cqa}{CQA}{critical quality attribute} \newacronym{cpp}{CPP}{critical process parameter} \newacronym{dms}{DMS}{degradable microscaffold} \newacronym{doe}{DOE}{design of experiments} \newacronym{adoe}{ADOE}{adaptive design of experiments} \newacronym{gmp}{GMP}{Good Manufacturing Practices} \newacronym{cho}{CHO}{Chinese hamster ovary} \newacronym{all}{ALL}{acute lymphoblastic leukemia} \newacronym{pdms}{PDMS}{polydimethylsiloxane} \newacronym{dc}{DC}{dendritic cell} \newacronym{il2}{IL2}{interleukin 2} \newacronym{il15}{IL15}{interleukin 15} \newacronym{il15r}{IL15R}{interleukin 15 receptor} \newacronym{rhil2}{rhIL2}{recombinant human interleukin 2} \newacronym{apc}{APC}{antigen presenting cell} \newacronym{mhc}{MHC}{major histocompatibility complex} \newacronym{elisa}{ELISA}{enzyme-linked immunosorbent assay} \newacronym{nmr}{NMR}{nuclear magnetic resonance} \newacronym{haba}{HABA}{4-hydroxyazobenene-2-carboxylic-acid} \newacronym{pbs}{PBS}{phosphate buffered saline} \newacronym{bca}{BCA}{bicinchoninic acid assay} \newacronym{bsa}{BSA}{bovine serum albumin} \newacronym{hsa}{HSA}{human serum albumin} \newacronym{stp}{STP}{streptavidin} \newacronym{stppe}{STP-PE}{streptavidin-phycoerythrin} \newacronym{snb}{SNB}{sulfo-nhs-biotin} \newacronym{cug}{CuG}{Cultispher G} \newacronym{cus}{CuS}{Cultispher S} \newacronym{pbmc}{PBMC}{peripheral blood mononuclear cells} \newacronym{macs}{MACS}{magnetic activated cell sorting} \newacronym{aopi}{AO/PI}{acridine orange/propidium iodide} \newacronym{igg}{IgG}{immunoglobulin G} \newacronym{pe}{PE}{phycoerythrin} \newacronym{fitc}{FITC}{Fluorescein} \newacronym{fitcbt}{FITC-BT}{Fluorescein-biotin} \newacronym{ptnl}{PTN-L}{Protein L} \newacronym{af647}{AF647}{Alexa Fluor 647} \newacronym{anova}{ANOVA}{analysis of variance} \newacronym{crispr}{CRISPR}{clustered regularly interspaced short palindromic repeats} \newacronym{mtt}{MTT}{3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide} \newacronym{bmi}{BMI}{body mass index} \newacronym{a2b1}{A2B1}{integrin $\upalpha$1$\upbeta$1} \newacronym{a2b2}{A2B2}{integrin $\upalpha$1$\upbeta$2} \newacronym{til}{TIL}{tumor infiltrating lymphocytes} \newacronym{nsg}{NSG}{NOD scid gamma} \newacronym{colb}{COL-B}{collagenase B} \newacronym{cold}{COL-D}{collagenase D} \newacronym{tsne}{tSNE}{t-stochastic neighbor embedding} \newacronym{anv}{AXV}{Annexin-V} \newacronym{pi}{PI}{propidium iodide} \newacronym{rt}{RT}{room temperature} \newacronym{cas37}{Cas3/7}{Caspase-3/7} \newacronym{bcl2}{BCL-2}{B cell lymphoma 2} \newacronym{tmb}{TMB}{3,3',5,5'-Tetramethylbenzidine} \newacronym{gvhd}{GVHD}{graft-vs-host disease} \newacronym{bcma}{BCMA}{B-cell maturation antigen} \newacronym{di}{DI}{deionized} \newacronym{moi}{MOI}{multiplicity of infection} \newacronym{ifng}{IFN$\upgamma$}{interferon-$\upgamma$} \newacronym{tnfa}{TNF$\upalpha$}{tumor necrosis factor-$\upalpha$} \newacronym{sql}{SQL}{structured query language} \newacronym{fcs}{FCS}{flow cytometry standard} \newacronym{ivis}{ivis}{in vivo imaging system} \newacronym{iacuc}{IACUC}{institutional animal care and use committee} \newacronym{hbss}{HBSS}{Hank's buffered saline solution} \newacronym{leaf}{LEAF}{low endotoxin, azide-free} \newacronym{cytof}{CyTOF}{cytometry time-of-flight} \newacronym{spade}{SPADE}{spanning-tree progression analysis of density-normalized events} %%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%% % SI units for uber nerds % NOTE the \SI macro is depreciated but the arch repo (!!!) hasn't been updated % with the latest package yet (texlive-science) \sisetup{per-mode=symbol,list-units=single} \DeclareSIUnit\IU{IU} \DeclareSIUnit\rpm{RPM} \DeclareSIUnit\carrier{carrier} \DeclareSIUnit\dms{DMS} \DeclareSIUnit\cell{cells} \DeclareSIUnit\ab{mAb} \DeclareSIUnit\normal{N} \DeclareSIUnit\molar{M} \DeclareSIUnit\mM{\milli\molar} \DeclareSIUnit\uM{\micro\molar} \DeclareSIUnit\gforce{\times{} g} %%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%% % commands for lazy farts like me \newcommand{\mytitle}{ \Large{ \textbf{ Optimizing T Cell Manufacturing and Quality Using Functionalized Degradable Microscaffolds } } } \newcommand{\mycommitteemember}[3]{ \begin{flushleft} \noindent #1 \\ #2 \\ \textit{#3} \end{flushleft} } % a BME's best friend \newcommand{\invivo}{\textit{in vivo}} \newcommand{\invitro}{\textit{in vitro}} \newcommand{\exvivo}{\textit{ex vivo}} % various CD-whatever crap \newcommand{\cd}[1]{CD{#1}} \newcommand{\anti}[1]{anti-{#1}} \newcommand{\antih}[1]{anti-human {#1}} \newcommand{\antim}[1]{anti-mouse {#1}} \newcommand{\acd}[1]{\anti{\cd{#1}}} \newcommand{\ahcd}[1]{\antih{\cd{#1}}} \newcommand{\amcd}[1]{\antim{\cd{#1}}} \newcommand{\pos}[1]{#1+} \newcommand{\cdp}[1]{\pos{\cd{#1}}} \newcommand{\cdn}[1]{\cd{#1}-} \newcommand{\ptmem}{\cdp{62L}\pos{CCR7}} \newcommand{\ptmemp}{\ptmem{}~\si{\percent}} \newcommand{\pth}{\cdp{4}} \newcommand{\pthp}{\pth{}~\si{\percent}} \newcommand{\ptk}{\cdp{8}} \newcommand{\ptmemh}{\pth\ptmem} \newcommand{\ptmemk}{\ptk\ptmem} \newcommand{\dpthp}{$\Updelta$\pthp{}} \newcommand{\ptcar}{\gls{car}+} \newcommand{\ptcarp}{\ptcar~\si{\percent}} % DOE responses I don't feel like typing ad-nauseam \newcommand{\pilII}{\gls{il2} concentration} \newcommand{\pdms}{\gls{dms} concentration} \newcommand{\pmab}{functional \gls{mab} surface density} % vendor and product stuff I don't feel like typing \newcommand{\catnum}[2]{(#1, #2)} \newcommand{\product}[3]{#1 \catnum{#2}{#3}} \newcommand{\thermo}{Thermo Fisher} \newcommand{\miltenyi}{Miltenyi Biotech} \newcommand{\bl}{Biolegend} % the obligatory misc category \newcommand{\inlinecode}{\texttt} \newcommand{\subcap}[2]{\subref{#1}) #2} \newcommand{\sigkey}{Significance test key: *p<0.1; **p < 0.05; ***p<0.01} %%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%% % ditto for environments \newenvironment{mytitlepage}{ \begin{singlespace} \begin{center} } { \end{center} \end{singlespace} } %%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%% % begin document (proceed with caution) \begin{document} \begin{titlepage} \begin{mytitlepage} \mytitle{} \vfill \Large{ A Dissertation \\ Presented to \\ The Academic Faculty \\ \vspace{1.5em} by \vspace{1.5em} Nathan John Dwarshuis, B.S. \\ \vfill In Partial Fulfillment \\ of the Requirements for the Degree \\ Doctor of Philosophy in Biomedical Engineering in the \\ Wallace H. Coulter Department of Biomedical Engineering \vfill Georgia Institute of Technology and Emory University \\ August 2021 \vfill COPYRIGHT \copyright{} BY NATHAN J. DWARSHUIS } \end{mytitlepage} \end{titlepage} \onecolumn \pagenumbering{roman} \clearpage \begin{mytitlepage} \mytitle{} \end{mytitlepage} \vfill \large{ \noindent Committee Members \begin{multicols}{2} \begin{singlespace} \mycommitteemember{Dr.\ Krishnendu\ Roy\ (Advisor)} {Department of Biomedical Engineering} {Georgia Institute of Technology and Emory University} \vspace{1.5em} \mycommitteemember{Dr.\ Madhav\ Dhodapkar} {Department of Hematology and Medical Oncology} {Emory University} \vspace{1.5em} \mycommitteemember{Dr.\ Melissa\ Kemp} {Department of Biomedical Engineering} {Georgia Institute of Technology and Emory University} \columnbreak{} \null{} \vfill \mycommitteemember{Dr.\ Wilbur\ Lam} {Department of Biomedical Engineering} {Georgia Institute of Technology and Emory University} \vspace{1.5em} \mycommitteemember{Dr.\ Sakis\ Mantalaris} {Department of Biomedical Engineering} {Georgia Institute of Technology and Emory University} \end{singlespace} \end{multicols} \vspace{1.5em} \hfill Date Approved: } \clearpage \chapter*{acknowledgements} \addcontentsline{toc}{chapter}{acknowledgements} Thank you to Lex Fridman and Devin Townsend for being awesome and inspirational. \clearpage \chapter*{summary} \addcontentsline{toc}{chapter}{summary} \Gls{act} using \gls{car} T cells have shown promise in treating cancer, but manufacturing large numbers of high quality cells remains challenging. Currently approved T cell expansion technologies involve \anti-cd{3} and \anti-cd{28} \glspl{mab}, usually mounted on magnetic beads. This method fails to recapitulate many key signals found \invivo{} and is also heavily licensed by a few companies, limiting its long-term usefulness to manufactures and clinicians. Furthermore, we understand that highly potent T cells are generally less-differentiated subtypes such as central memory and stem memory T cells. Despite this understanding, little has been done to optimize T cell expansion for generating these subtypes, including measurement and feedback control strategies that are necessary for any modern manufacturing process. The goal of this thesis was to develop a microcarrier-based \gls{dms} T cell expansion system as well as determine biologically-meaningful \glspl{cqa} and \glspl{cpp} that could be used to optimize for highly-potent T cells. In Aim 1, we develop and characterized the \gls{dms} system, including quality control steps. We also demonstrate the feasiblity of expanding highly-potent memory and CD4+ T cells, and showing compatibility with existing \gls{car} transduction methods. In aim 2, we use \gls{doe} methodology to optimize the \gls{dms} platform, and develop a computational pipeline to identify and model the effect of measurable \glspl{cqa} and \glspl{cpp} on the final product. In aim 3, we demonstrate the effectiveness of the \gls{dms} platform \invivo{}. This thesis lays the groundwork for a novel T cell expansion method which can be used in a clinical setting, and also provides a path toward optimizing for product quality in an industrial setting. \clearpage \tableofcontents \clearpage \listoffigures \clearpage \listoftables \clearpage % \twocolumn \chapter*{acronyms} \addcontentsline{toc}{chapter}{acronyms} \printglossary[type=\acronymtype] \clearpage \pagenumbering{arabic} \clearpage \chapter{introduction} \section*{overview} % TODO this is basically the same as the first part of the backgound, I guess I % can just trim it down T cell-based immunotherapies have received great interest from clinicians and industry due to their potential to treat, and often cure, cancer and other diseases\cite{Fesnak2016,Rosenberg2015}. In 2017, Novartis and Kite Pharma received FDA approval for \textit{Kymriah} and \textit{Yescarta} respectively, two genetically-modified \gls{car} T cell therapies against B cell malignancies. Despite these successes, \gls{car} T cell therapies are constrained by an expensive and difficult-to-scale manufacturing process with little control on cell quality and phenotype3,4. State-of-the-art T cell manufacturing techniques focus on \acd{3} and \acd{28} activation and expansion, typically presented on superparamagnetic, iron-based microbeads (Invitrogen Dynabead, Miltenyi MACS beads), on nanobeads (Miltenyi TransACT), or in soluble tetramers (Expamer)\cite{Roddie2019,Dwarshuis2017,Wang2016, Piscopo2017, Bashour2015}. These strategies overlook many of the signaling components present in the secondary lymphoid organs where T cells expand \invivo{}. Typically, T cells are activated under close cell-cell contact, which allows for efficient autocrine/paracrine signaling via growth-stimulating cytokines such as \gls{il2}. Additionally, the lymphoid tissues are comprised of \gls{ecm} components such as collagen, which provide signals to upregulate proliferation, cytokine production, and pro-survival pathways\cite{Gendron2003, Ohtani2008, Boisvert2007, Ben-Horin2004}. We hypothesized that culture conditions that better mimic these \invivo{} expansion conditions of T cells, can significantly improve the quality and quantity of manufactured T cells and provide better control on the resulting T cell phenotype. % TODO mention the Cloudz stuff that's in my presentation A variety of solutions have been proposed to make the T cell expansion process more physiological. One strategy is to use modified feeder cell cultures to provide activation signals similar to those of \glspl{dc}\cite{Forget2014}. While this has the theoretical capacity to mimic many components of the lymph node, it is hard to reproduce on a large scale due to the complexity and inherent variability of using cell lines in a fully \gls{gmp}-compliant manner. Others have proposed biomaterials-based solutions to circumvent this problem, including lipid-coated microrods\cite{Cheung2018}, 3D-scaffolds via either Matrigel\cite{Rio2018} or 3d-printed lattices\cite{Delalat2017}, ellipsoid beads\cite{meyer15_immun}, and \gls{mab}-conjugated \gls{pdms} beads\cite{Lambert2017} that respectively recapitulate the cellular membrane, large interfacial contact area, 3D-structure, or soft surfaces T cells normally experience \invivo{}. While these have been shown to provide superior expansion compared to traditional microbeads, none of these methods has been able to show preferential expansion of functional naïve/memory and CD4 T cell populations. Generally, T cells with a lower differentiation state such as naïve and memory cells have been shown to provide superior anti-tumor potency, presumably due to their higher potential to replicate, migrate, and engraft, leading to a long-term, durable response\cite{Xu2014, Fraietta2018, Gattinoni2011, Gattinoni2012}. Likewise, CD4 T cells are similarly important to anti-tumor potency due to their cytokine release properties and ability to resist exhaustion\cite{Wang2018, Yang2017}. Therefore, methods to increase naïve/memory and CD4 T cells in the final product are needed, a critical consideration being ease of translation to industry and ability to interface with scalable systems such as bioreactors. % TODO probably need to address some of the modeling stuff here as well This thesis describes a novel degradable microscaffold-based method derived from porous microcarriers functionalized with \acd{3} and \acd{28} \glspl{mab} for use in T cell expansion cultures. Microcarriers have historically been used throughout the bioprocess industry for adherent cultures such as stem cells and \gls{cho} cells, but not with suspension cells such as T cells\cite{Heathman2015, Sart2011}. The microcarriers chosen to make the DMSs in this study have a microporous structure that allows T cells to grow inside and along the surface, providing ample cell-cell contact for enhanced autocrine and paracrine signaling. Furthermore, the carriers are composed of gelatin, which is a collagen derivative and therefore has adhesion domains that are also present within the lymph nodes. Finally, the 3D surface of the carriers provides a larger contact area for T cells to interact with the \glspl{mab} relative to beads; this may better emulate the large contact surface area that occurs between T cells and \glspl{dc}. These microcarriers are readily available in over 30 countries and are used in an FDA fast-track-approved combination retinal pigment epithelial cell product (Spheramine, Titan Pharmaceuticals) {\#}[Purcell documentation]. This regulatory history will aid in clinical translation. We show that compared to traditional microbeads, \gls{dms}-expanded T cells not only provide superior expansion, but consistently provide a higher frequency of naïve/memory and CD4 T cells (CCR7+CD62L+) across multiple donors. We also demonstrate functional cytotoxicity using a CD19 \gls{car} and a superior performance, even at a lower \gls{car} T cell dose, of \gls{dms}-expanded \gls{car}-T cells \invivo{} in a mouse xenograft model of human B cell \gls{all}. Our results indicate that \glspl{dms} provide a robust and scalable platform for manufacturing therapeutic T cells with higher naïve/memory phenotype and more balanced CD4+ T cell content. \section*{hypothesis} The hypothesis of this dissertation was that using \glspl{dms} created from off-the-shelf microcarriers and coated with activating \glspl{mab} would lead to higher quantity and quality T cells as compared to state-of-the-art bead-based expansion. The objective of this dissertation was to develop this platform, test its effectiveness both \invivo{} and \invivo{}, and develop computational pipelines that could be used in a manufacturing environment. \section*{specific aims} The specific aims of this dissertation are outlined in \cref{fig:graphical_overview}. \begin{figure*}[ht!] \begingroup \includegraphics[width=\textwidth]{../figures/overview.png} \endgroup \caption[Project Overview]{High-level overview.} \label{fig:graphical_overview} \end{figure*} \subsection*{aim 1: develop and optimize a novel T cell expansion process that mimics key components of the lymph nodes} % TODO this might be easier to break apart in separate aims In this first aim, we demonstrated the process for manufacturing \glspl{dms}, including quality control steps that are necessary for translation of this platform into a scalable manufacturing setting. We also demonstrate that the \gls{dms} platform leads to higher overall expansion of T cells and higher overall fractions of potent memory and CD4+ subtypes desired for T cell therapies. Finally, we demonstrate \invitro{} that the \gls{dms} platform can be used to generate functional \gls{car} T cells targeted toward CD19. \subsection*{aim 2: develop methods to control and predict T cell quality} For this second aim, we investigated methods to identify and control \glspl{cqa} and glspl{cpp} for manufacturing T cells using the \gls{dms} platform. This was accomplished through two sub-aims: \begin{itemize} \item[A --] Develop computational methods to control and predict T cell expansion and quality \item[B --] Perturb \gls{dms} expansion to identify additional mechanistic controls for expansion and quality \end{itemize} \subsection*{aim 3: confirm potency of T cells from novel T cell expansion process using \invivo{} xenograft mouse model} In this final aim, we demonstrate the effectiveness of \gls{dms}-expanded T cells compared to state-of-the-art beads using \invivo{} mouse models for \gls{all}. \section*{outline} In Chapter~\ref{background}, we provide additional background on the current state of T cell manufacturing and how the work in this dissertation moves the field forward. In Chapters~\ref{aim1},~\ref{aim2a},~\ref{aim2b}, and~\ref{aim3} we present the work pertaining to Aims 1, 2, and 3 respectively. Finally, we present our final conclusions in Chapter~\ref{conclusions}. \chapter{background and significance}\label{background} \section*{background} % TODO break this apart into mfg tech and T cell phenotypes/quality % TODO consider adding a separate section on microcarriers and their use in % bioprocess % TODO add stuff about T cell licensing \subsection*{current T cell manufacturing technologies} \Gls{car} T cell therapy has received great interest from both academia and industry due to its potential to treat cancer and other diseases\cite{Fesnak2016, Rosenberg2015}. In 2017, Novartis and Kite Pharma acquired FDA approval for \textit{Kymriah} and \textit{Yescarta} respectively, two \gls{car} T cell therapies against B cell malignancies. Despite these successes, \gls{car} T cell therapies are constrained by an expensive and difficult-to-scale manufacturing process\cite{Roddie2019, Dwarshuis2017}. Of critical concern, state-of-the-art manufacturing techniques focus only on Signal 1 and Signal 2-based activation via \acd{3} and \acd{28} \glspl{mab}, typically presented on a microbead (Invitrogen Dynabead, Miltenyi MACS beads) or nanobead (Miltenyi TransACT), but also in soluble forms in the case of antibody tetramers (Expamer)\cite{Wang2016, Piscopo2017, Roddie2019, Bashour2015}. These strategies overlook many of the signaling components present in the secondary lymphoid organs where T cells normally expand. Typically, T cells are activated under close cell-cell contact via \glspl{apc} such as \glspl{dc}, which present peptide-\glspl{mhc} to T cells as well as a variety of other costimulatory signals. These close quarters allow for efficient autocrine/paracrine signaling among the expanding T cells, which secrete gls{il2} and other cytokines to assist their own growth. Additionally, the lymphoid tissues are comprised of \gls{ecm} components such as collagen, which provide signals to upregulate proliferation, cytokine production, and pro-survival pathways\cite{Gendron2003, Ohtani2008, Boisvert2007, Ben-Horin2004}. A variety of solutions have been proposed to make the T cell expansion process more physiological. One strategy is to use modified feeder cell cultures to provide activation signals similar to those of \glspl{dc}\cite{Forget2014}. While this has the theoretical capacity to mimic several key components of the lymph node, it is hard to reproduce on a large scale due to the complexity and inherent variability of using cell lines in a fully \gls{gmp}-compliant manner. Others have proposed biomaterials-based solutions to circumvent this problem, including lipid-coated microrods\cite{Cheung2018}, 3D-scaffolds via either Matrigel\cite{Rio2018} or 3d-printed lattices\cite{Delalat2017}, ellipsoid beads\cite{meyer15_immun}, and \gls{mab}-conjugated \gls{pdms} beads\cite{Lambert2017} that respectively recapitulate the cellular membrane, large interfacial contact area, 3D-structure, or soft surfaces T cells normally experience \textit{in vivo}. While these have been shown to provide superior expansion compared to traditional microbeads, no method has been able to show preferential expansion of functional memory and CD4 T cell populations. Generally, T cells with a lower differentiation state such as memory cells have been shown to provide superior anti-tumor potency, presumably due to their higher potential to replicate, migrate, and engraft, leading to a long-term, durable response\cite{Xu2014, Gattinoni2012, Fraietta2018, Gattinoni2011}. Likewise, CD4 T cells are similarly important to anti-tumor potency due to their cytokine release properties and ability to resist exhaustion\cite{Wang2018, Yang2017}, and no method exists to preferentially expand the CD4 population compared to state-of-the-art systems. Here we propose a method using microcarriers functionalized with \acd{3} and \acd{28} \glspl{mab} for use in T cell expansion cultures. Microcarriers have historically been used throughout the bioprocess industry for adherent cultures such as stem cells and \gls{cho} cells, but not with suspension cells such as T cells\cite{Heathman2015, Sart2011}. The carriers have a macroporous structure that allows T cells to grow inside and along the surface, providing ample cell-cell contact for enhanced autocrine and paracrine signaling. Furthermore, the carriers are composed of gelatin, which is a collagen derivative and therefore has adhesion domains that are also present within the lymph nodes. Finally, the 3D surface of the carriers provides a larger contact area for T cells to interact with the \glspl{mab} relative to beads; this may better emulate the large contact surface area that occurs between T cells and \glspl{dc}. \subsection*{strategies to optimize cell manufacturing} The \gls{dms} system has a number of parameters that can be optimized, and a \gls{doe} is an ideal framework to test multiple parameters simultaneously. The goal of \gls{doe} is to answer a data-driven question with the least number of resources. It was developed in many non-biological industries throughout the \nth{20} century such as the automotive and semiconductor industries where engineers needed to minimize downtime and resource consumption on full-scale production lines. % TODO add a bit more about the math of a DOE here \Glspl{doe} served three purposes in this dissertation. First, we used them as screening tools, which allowed us to test many input parameters and filter out the few that likely have the greatest effect on the response. Second, they were used to make a robust response surface model to predict optimums using relatively few resources, especially compared to full factorial or one-factor-at-a-time approaches. Third, we used \glspl{doe} to discover novel effects and interactions that generated hypotheses that could influence the directions for future work. \subsection*{strategies to characterize cell manufacturing} A number of multiomics strategies exist which can generate rich datasets for T cells. We will consider several multiomics strategies within this proposal: \begin{description} \item[Luminex:] A multiplexed bead-based \gls{elisa} that can measure many bulk (not single cell) cytokine concentrations simultaneously in a media sample. Since this only requires media (as opposed to destructively measuring cells) we will use this as a longitudinal measurement. \item[Metabolomics:] It is well known that T cells of different lineages have different metabolic profiles; for instance memory T cells have larger aerobic capacity and fatty acid oxidation\cite{Buck2016, van_der_Windt_2012}. We will interrogate key metabolic species using \gls{nmr} in collaboration with the Edison Lab at the University of Georgia. This will be both a longitudinal assay using media samples (since some metabolites may be expelled from cells that are indicative of their phenotype) and at endpoint where we will lyse the cells and interogate their entire metabolome. \item[Flow and Mass Cytometry:] Flow cytometry using fluorophores has been used extensively for immune cell analysis, but has a practical limit of approximately 18 colors\cite{Spitzer2016}. Mass cytometry is analogous to traditional flow cytometry except that it uses heavy-metal \gls{mab} conjugates, which has a practical limit of over 50 markers. This will be useful in determining precise subpopulations and phenotypes that may be influencing responses, especially when one considers that many cell types can be defined by more than one marker combination. We will perform this at endpoint. While mass cytometry is less practical than simple flow cytometers such as the BD Accuri, we may find that only a few markers are required to accurately predict performance, and thus this could easily translate to industry using relatively cost-effective equipment. \end{description} % TODO add a computational section % TODO add a section explaining causal inference since this is a big part of % the end of aim 1 \section{Innovation} \subsection{Innovation} Several aspects of this work are novel considering the state-of-the-art technology for T cell manufacturing: \begin{itemize} \item \Glspl{dms} offers a compelling alternative to state-of-the-art magnetic bead technologies (e.g. DynaBeads, MACS-Beads), which is noteworthy because the licenses for these techniques is controlled by only a few companies (Invitrogen and Miltenyi respectively). Because of this, bead-based expansion is more expensive to implement and therefore hinders companies from entering the rapidly growing T cell manufacturing arena. Providing an alternative as we are doing will add more options, increase competition among both raw material and T cell manufacturers, and consequently drive down cell therapy market prices and increase innovation throughout the industry. \item This is the first technology for T cell immunotherapies that selectively expands memory T cell populations with greater efficiency relative to bead-based expansion Others have demonstrated methods that can achieve greater expansion of T cells, but not necessarily specific populations that are known to be potent. \item We propose to optimize our systems using \gls{doe} methodology, which is a strategy commonly used in other industries and disciplines but has yet to gain wide usage in the development of cell therapies. \Glspl{doe} are advantageous as they allow the inspection of multiple parameters simultaneously, allowing efficient and comprehensive analysis of the system vs a one-factor-at-a-time approach. We believe this method is highly relevant to the development of cell therapies, not only for process optimization but also hypotheses generation. Of further note, most \textit{in vivo} experiments are not done using a \gls{doe}-based approach; however, a \gls{doe} is perfectly natural for a large mouse study where one naturally desires to use as few animals as possible. \item The \gls{dms} system is be compatible with static bioreactors such as the G-Rex which has been adopted throughout the cell therapy industry. Thus this technology can be easily incorporated into existing cell therapy process that are performed at scale. \item We analyzed our system using a multiomics approach, which will enable the discovery of novel biomarkers to be used as \glspl{cqa}. While this approach has been applied to T cells previously, it has not been done in the context of a large \gls{doe}-based model. This approach is aware of the whole design space, and thus enables greater understanding of process parameters and their effect on cell phenotype. \end{itemize} \chapter{aim 1}\label{aim1} \section{introduction} The first aim was to develop a microcarrier system that mimics several key aspects of the \invivo{} lymph node microenvironment. We compared compare this system to state-of-the-art T cell activation technologies for both expansion potential and memory cell formation. The governing hypothesis was that microcarriers functionalized with \acd{3} and \acd{28} \glspl{mab} will provide superior expansion and memory phenotype compared to state-of-the-art bead-based T cell expansion technology. % TODO this doesn't flow that well and is repetitive with what comes above Microcarriers have been used throughout the bioprocess industry for adherent cell cultures such as \gls{cho} cells and stem cells, as they are able to achieve much greater surface area per unit volume than traditional 2D cultures\cite{Heathman2015, Sart2011}. Adding adhesive \glspl{mab} to the microcarriers will adapt them for suspension cell cultures such as T cells. Consequently, the large macroporous structure will allow T cells to cluster more closely, which in turn will enable better autocrine and paracrine signaling. Specifically, two cytokines that are secreted by T cells, IL-2 and IL-15, are known to drive expansion and memory phenotype respectively\cite{Buck2016}. Therefore, the proposed microcarrier system should enable greater expansion and better retention of memory phenotype compared to current bead-based methods. \section{methods} \subsection{dms functionalization}\label{sec:dms_fab} \begin{figure*}[ht!] \begingroup \includegraphics{../figures/dms_flowchart.png} \endgroup \caption[\gls{dms} Flowchart]{Overview of \gls{dms} manufacturing process.} \label{fig:dms_flowchart} \end{figure*} Gelatin microcarriers (\gls{cus} or \gls{cug}, GE Healthcare, DG-2001-OO and DG-0001-OO) were suspended at \SI{20}{\mg\per\ml} in 1X \gls{pbs} and autoclaved. All subsequent steps were done aseptically, and all reactions were carried out at \SI{20}{\mg\per\ml} carriers at room temperature and agitated using an orbital shaker with a \SI{3}{\mm} orbit diameter. After autoclaving, the microcarriers were washed using sterile \gls{pbs} three times in a 10:1 volume ratio. \product{\Gls{snb}}{\thermo}{21217} was dissolved at approximately \SI{10}{\uM} in sterile ultrapure water, and the true concentration was then determined using the \gls{haba} assay (see below). \SI{5}{\ul\of{\ab}\per\mL} \gls{pbs} was added to carrier suspension and allowed to react for \SI{60}{\minute} at \SI{700}{\rpm} of agitation. After the reaction, the amount of biotin remaining in solution was quantified using the \gls{haba} assay (see below). The carriers were then washed three times, which entailed adding sterile \gls{pbs} in a 10:1 volumetric ratio, agitating at \SI{900}{\rpm} for \SI{10}{\minute}, adding up to a 15:1 volumetric ratio (relative to reaction volume) of sterile \gls{pbs}, centrifuging at \SI{1000}{\gforce} for \SI{1}{\minute}, and removing all liquid back down to the reaction volume. To coat with \gls{stp}, \SI{40}{\ug\per\mL} \product{\gls{stp}}{Jackson Immunoresearch}{016-000-114} was added and allowed to react for \SI{60}{\minute} at \SI{700}{RPM} of agitation. After the reaction, supernatant was taken for the \product{\gls{bca} assay}{\thermo}{23225}, and the carriers were washed analogously to the previous wash step to remove the biotin, except two washes were done and the agitation time was \SI{30}{\minute}. Biotinylated \glspl{mab} against human CD3 \catnum{\bl}{317320} and CD28 \catnum{\bl}{302904} were combined in a 1:1 mass ratio and added to the carriers at \SI{0.2}{\ug\of{\ab}\per\mg\of{\dms}}. Along with the \glspl{mab}, sterile \product{\gls{bsa}}{Sigma}{A9576} was added to a final concentration of \SI{2}{\percent} in order to prevent non-specific binding of the antibodies to the reaction tubes. \glspl{mab} were allowed to bind to the carriers for \SI{60}{\minute} with \SI{700}{\rpm} agitation. After binding, supernatants were sampled to quantify remaining \gls{mab} concentration using an \product{\anti{\gls{igg}} \gls{elisa} kit}{Abcam}{157719}. Fully functionalized \glspl{dms} were washed in sterile \gls{pbs} analogous to the previous washing step to remove excess \gls{stp}. They were washed once again in the cell culture media to be used for the T cell expansion. The concentration of the final \gls{dms} suspension was found by taking a \SI{50}{\uL} sample, plating in a well, and imaging the entire well. The image was then manually counted to obtain a concentration. Surface area for \si{\ab\per\um\squared} was calculated using the properties for \gls{cus} and \gls{cug} as given by the manufacturer {Table X}. \subsection{dms quality control assays} Biotin was quantified using the \product{\gls{haba} assay}{Sigma}{H2153-1VL}. In the case of quantifying \gls{snb} prior to adding it to the microcarriers, the sample volume was quenched in a 1:1 volumetric ratio with \SI{1}{\molar} NaOH and allowed to react for \SI{1}{\minute} in order to prevent the reactive ester linkages from binding to the avidin proteins in the \gls{haba}/avidin premix. All quantifications of \gls{haba} were performed on an Eppendorf D30 Spectrophotometer using \product{\SI{70}{\ul} cuvettes}{BrandTech}{759200}. The extinction coefficient at \SI{500}{\nm} for \gls{haba}/avidin was assumed to be \SI{34000}{\per\cm\per\molar}. \gls{stp} binding to the carriers was quantified indirectly using a \product{\gls{bca} kit}{\thermo}{23227} according to the manufacturer’s instructions, with the exception that the standard curve was made with known concentrations of purified \gls{stp} instead of \gls{bsa}. Absorbance at \SI{592}{\nm} was quantified using a Biotek plate reader. \Gls{mab} binding to the microcarriers was quantified indirectly using an \gls{elisa} assay per the manufacturer’s instructions, with the exception that the same antibodies used to coat the carriers were used as the standard for the \gls{elisa} standard curve. Open biotin binding sites on the \glspl{dms} after \gls{stp} coating was quantified indirectly using \product{\gls{fitcbt}}{\thermo}{B10570}. Briefly, \SI{400}{\pmol\per\ml} \gls{fitcbt} were added to \gls{stp}-coated carriers and allowed to react for \SI{20}{\minute} at room temperature under constant agitation. The supernatant was quantified against a standard curve of \gls{fitcbt} using a Biotek plate reader. \Gls{stp} binding was verified after the \gls{stp}-binding step visually by adding \gls{fitcbt} to the \gls{stp}-coated \glspl{dms}, resuspending in \SI{1}{\percent} agarose gel, and imaging on a \product{lightsheet microscope}{Zeiss}{Z.1}. \Gls{mab} binding was verified visually by first staining with \product{\anti{\gls{igg}}-\gls{fitc}}{\bl}{406001}, incubating for \SI{30}{\minute}, washing with \gls{pbs}, and imaging on a confocal microscope. \subsection{t cell culture}\label{sec:tcellculture} % TODO verify countess product number Cryopreserved primary human T cells were either obtained as sorted \product{\cdp{3} T cells}{Astarte Biotech}{1017} or isolated from \product{\glspl{pbmc}}{Zenbio}{SER-PBMC} using a negative selection \product{\cdp{3} \gls{macs} kit}{\miltenyi}{130-096-535}. T cells were activated using \glspl{dms} or \product{\SI{3.5}{\um} CD3/CD28 magnetic beads}{\miltenyi}{130-091-441}. In the case of beads, T cells were activated at the manufacturer recommended cell:bead ratio of 2:1. In the case of \glspl{dms}, cells were activated using \SI{2500}{\dms\per\cm\squared} unless otherwise noted. Initial cell density was \SIrange{2e6}{2.5e6}{\cell\per\ml} to in a 96 well plate with \SI{300}{\ul} volume. Serum-free media was either \product{OpTmizer}{\thermo}{A1048501} or \product{TexMACS}{\miltenyi}{170-076-307} supplemented with \SIrange{100}{400}{\IU\per\ml} \product{\gls{rhil2}}{Peprotech}{200-02}. Cell cultures were expanded for \SI{14}{\day} as counted from the time of initial seeding and activation. Cell counts and viability were assessed using \product{trypan blue}{\thermo}{T10282} or \product{\gls{aopi}}{Nexcelom Bioscience}{CS2-0106-5} and a \product{Countess Automated Cell Counter}{Thermo Fisher}{Countess 3 FL}. Media was added to cultures every \SIrange{2}{3}{\day} depending on media color or a \SI{300}{\mg\per\deci\liter} minimum glucose threshold. Media glucose was measured using a \product{GlucCell glucose meter}{Chemglass}{CLS-1322-02}. Cells on the \glspl{dms} were visualized by adding \SI{0.5}{\ul} \product{\gls{stppe}}{\bl}{405204} and \SI{2}{ul} \product{\acd{45}-\gls{af647}}{\bl}{368538}, incubating for \SI{1}{\hour}, and imaging on a spinning disk confocal microscope. In the case of Grex bioreactors, we either used a \product{24 well plate}{Wilson Wolf}{P/N 80192M} or a \product{6 well plate}{P/N 80240M}. \subsection{Quantifying cells on DMS interior} % TODO add a product number to MTT (if I can find it) Cells were stained and visualized using \gls{mtt}. \glspl{dms} with attached and loosely attached cells were sampled as desired and filtered through a \SI{40}{\um} cell strainer. While still in the cell strainer, \glspl{dms} were washed twice with \gls{pbs} and then dried by pulling liquid through the bottom of the cell strainer via a micropipette and dabbing with a KimWipe. \glspl{dms} were transferred to a 24 well plate with \SI{400}{\ul} media. \SI{40}{\ul} \gls{mtt} was added to each well and allowed to incubate for \SI{3}{\hour}, after which \glspl{dms} with cell were visualized via a brightfield microscope. To quantify cells on the interior of \glspl{dms}, cells and \glspl{dms} were isolated analogously to those for the \gls{mtt} stain up until the drying step. Cells were then transferred to a tube containing \SI{400}{\ul} at \SI{5}{\mg\per\ml} dispase solution. \glspl{dms} were incubated and rotated for \SI{45}{\minute} at \SI{37}{\degreeCelsius}, after which cells were counted as already described in \cref{sec:tcellculture}. \subsection{quantification of apoptosis using Annexin-V} Apoptosis was quantified using \gls{anv} according to the manufacturer's instructions. Briefly, cells were transferred to flow tubes and washed twice with \gls{pbs} by adding \SI{3}{\ml} to each tube, centrifuging for \SI{400}{\gforce}, and aspirating the liquid down to \SI{200}{\ul}. The cells were analogously washed a third time with staining buffer (\SI{10}{\mM} HEPES, \SI{140}{\mM} NaCl, \SI{2.5}{\mM} CaCl\textsubscript{2}) and aspirated down to a final volume of \SI{100}{\ul}. Cells were stained in this volume with \SI{1}{\ul} \product{\gls{anv}-\gls{fitc}}{\bl}{640906} and \SI{5}{\ul} \product{\gls{pi}}{\thermo}{P3566} and incubated for \SI{15}{\minute} at gls{rt} in the dark. After incubation, \SI{400}{\ul} staining buffer was added to each tube. Each tube was then analyzed via flow cytometry. \subsection{quantification of Caspase-3/7} \Gls{cas37} was quantified using \product{CellEvent dye}{\thermo}{C10723} according the manufacturer's instructions. Briefly, a 2X (\SI{8}{\mM}) working solution of CellEvent dye was added to \SI{100}{\ul} cell suspension (at least \num{5e4} cells) and incubated at \SI{37}{\degreeCelsius} for \SI{30}{\minute}. After incubation, cells were immediately analyzed via flow cytometry. \subsection{quantification of BCL-2} \Gls{bcl2} was quantified using an \product{Human Total Bcl-2 DuoSet \gls{elisa} kit}{Rnd Systems}{DYC827B-2} according to the manufacturer's instructions and supplemented with \product{5X diluent buffer}{\bl}{421203}, \product{\gls{tmb} substrate solution}{eBioscience}{00-4201-56}, and \SI{2}{\normal} H\textsubscript{2}SO\textsubscript{4} stop solution made in house. Briefly, cells were lysed using \product{10X lysis buffer}{Cell Signaling}{9803S}, and the lysate was quantified for protein using a \product{\gls{bca} assay}{\thermo}{23225} as directed. Standardized lysates were measured using the \gls{elisa} kit as directed. \subsection{chemotaxis assay} % TODO not sure about the transwell product number Migratory function was assayed using a transwell chemotaxis assay as previously described\cite{Hromas1997}. Briefly, \SI{3e5}{\cell} were loaded into a \product{transwell plate with \SI{5}{\um} pore size}{Corning}{3421} with the basolateral chamber loaded with \SI{600}{\ul} media and 0, 250, or \SI{1000}{\ng\per\mL} \product{CCL21}{Peprotech}{250-13}. The plate was incubated for \SI{4}{\hour} after loading, and the basolateral chamber of each transwell was quantified for total cells using \product{countbright beads}{\thermo}{C36950}. The final readout was normalized using the \SI{0}{\ng\per\mL} concentration as background. \subsection{degranulation assay} Cytotoxicity of expanded \gls{car} T cells was assessed using a degranulation assay as previously described\cite{Schmoldt1975}. Briefly, \num{3e5} T cells were incubated with \num{1.5e5} target cells consisting of either \product{K562 wild type cells}{ATCC}{CCL-243} or CD19- expressing K562 cells transformed with \gls{crispr} (kindly provided by Dr.\ Yvonne Chen, UCLA)\cite{Zah2016}. Cells were seeded in a flat bottom 96 well plate with \SI{1}{\ug\per\ml} \product{\acd{49d}}{eBioscience}{16-0499-81}, \SI{2}{\micro\molar} \product{monensin}{eBioscience}{ 00-4505-51}, and \SI{1}{\ug\per\ml} \product{\acd{28}}{eBioscience}{302914} (all functional grade \glspl{mab}) with \SI{250}{\ul} total volume. After \SI{4}{\hour} incubation at \SI{37}{\degreeCelsius}, cells were stained for CD3, CD4, and CD107a and analyzed on a BD LSR Fortessa. Readout was calculated as the percent \cdp{107a} cells of the total \cdp{8} fraction. \subsection{car expression} \gls{car} expression was quantified as previously described\cite{Zheng2012}. Briefly, cells were washed once and stained with \product{biotinylated \gls{ptnl}}{\thermo}{29997}. After a subsequent wash, cells were stained with \product{\gls{pe}-\gls{stp}}{\bl}{405204}, washed again, and analyzed on a BD Accuri. Readout was percent \gls{pe}+ cells as compared to secondary controls (\gls{pe}-\gls{stp} with no \gls{ptnl}). % TODO add BCMA-CAR stuff \subsection{car plasmid and lentiviral transduction} The anti-CD19-CD8-CD137-CD3z \gls{car} with the EF1$\upalpha$ promotor\cite{Milone2009} was synthesized (Aldevron) and subcloned into a \product{FUGW}{Addgene}{14883} kindly provided by the Emory Viral Vector Core. Lentiviral vectors were synthesized by the Emory Viral Vector Core or the Cincinnati Children's Hospital Medical Center Viral Vector Core. To transduce primary human T cells, \product{retronectin}{Takara}{T100A} was coated onto non-TC treated 96 well plates and used to immobilize lentiviral vector particles according to the manufacturer's instructions. Briefly, retronectin solution was adsorbed overnight at \SI{4}{\degreeCelsius} and blocked the next day using \gls{bsa}. Prior to transduction, lentiviral supernatant was spinoculated at \SI{2000}{\gforce} for \SI{2}{\hour} at \SI{4}{\degreeCelsius}. T cells were activated in 96 well plates using beads or \glspl{dms} for \SI{24}{\hour}, and then cells and beads/\glspl{dms} were transferred onto lentiviral vector coated plates and incubated for another \SI{24}{\hour}. Cells and beads/\glspl{dms} were removed from the retronectin plates using vigorous pipetting and transferred to another 96 well plate wherein expansion continued. \subsection{sulfo-NHS-biotin hydrolysis quantification} The equation for hydrolysis of \gls{snb} was assumed to follow \cref{chem:snb_hydrolysis}. % TODO make this look prettier \begin{equation} \label{chem:snb_hydrolysis} \ce{NHS-CO-Biotin + OH- -> NHS- + Biotin-COOH} \end{equation} The hydrolysis of \gls{snb} was performed spectroscopically. \gls{snb} was added to either \gls{di} water or \gls{pbs} in a UV-transparent 96 well plate. Kinetic analysis using a Biotech Plate Reader began immediately after prep, and readings at \SI{260}{\nm} were taken every minute for \SI{2}{\hour}. \subsection{reaction kinetics quantification} The diffusion of \gls{stp} into biotin-coated microcarriers was determined experimentally. \SI{40}{\ug\per\ml} \gls{stp} was added to multiple batches of biotin-coated microcarriers, and supernatents were taken at fixed intervals and quantified for \gls{stp} protein using the \gls{bca} assay. % TODO defend why the microcarriers were saturated with stp The effective diffusivity of the microcarriers was determined using a pseudo-steady-state model. Each microcarrier was assumed to be a porous sphere with a fixed number of `\gls{stp} binding sites' equal to the maximum number of \gls{stp} molecules that could binding to the surface per area (eg, we assumed the surface was fully covered by \gls{stp}). Because the reaction rate between biotin and \gls{stp} was so fast, we assumed that the interface of free biotin shrunk as a function of \gls{stp} bound until the center of the microcarriers was reached. We also assumed that the pores in the microcarriers were large enough that the interactions between the \gls{stp} and surfaces would be small, thus the apparent diffusivity could be represented as a fraction of the diffusion coefficient of \gls{stp} in water. This model was given by \cref{eqn:stp_diffision_1,eqn:stp_diffision_2}: % TODO actually derive these equations, eg state the initial conditions and % governing equation \begin{equation} \label{eqn:stp_diffision_1} \frac{dr}{dt} = \frac{-D_{app}C}{Br(1-r/R)} \end{equation} \begin{equation} \label{eqn:stp_diffision_2} \frac{dC}{dt} = \frac{-4 \pi n D_{app} C}{V(1/r-1/R)} \end{equation} \noindent where \begin{itemize}[label={}] \item $D_{app}$ is the apparent diffusion rate which is equal to $D_{STP}\beta$ \item $D_{STP}$ the diffusion rate of \gls{stp} in water \item $\beta$ a fractional parameter representing the tortuousity and void fraction of the microcarriers. \item $r$ is the interfatial radius of the unbound biotin within a microcarrier \item $t$ is the reaction time \item $C$ is the concentration of \gls{stp} in the bulk solution \item $V$ is the volume of the bulk medium \item $R$ is the average radius of the microcarriers \item $n$ is the number of microcarriers in the reaction volume \end{itemize} % TODO cite the diffusion rate of stp The diffusion rate of \gls{stp} was assumed to be \SI{3.89e-7}{\cm\squared\per\second} {\#}{diffusion rate citation}. Since all but $\beta$ was known, the experimental data was fit using these equations using \inlinecode{ode45} in MATLAB and least squares as the fitting error. % TODO this diffusion rate isn't actually reflected in the code These equations were then used analogously to describe the reaction profile of \glspl{mab} assuming a diffusion rate of \SI{4.8e-7}{\cm\squared\per\second}\cite{Sherwood1992}. % METHOD add the equation governing the washing steps \subsection{Luminex Analysis}\label{sec:luminex_analysis} Luminex was performed using a \product{ProcartaPlex kit}{\thermo}{custom} for the markers outlined in \cref{tab:luminex_panel} with modifications (note that some markers were run in separate panels to allow for proper dilutions). Briefly, media supernatents from cells were sampled as desired and immediately placed in a \SI{-80}{\degreeCelsius} freezer until use. Before use, samples were thawed at \gls{rt} and vortexed to ensure homogeneity. To run the plate, \SI{25}{\ul} of magnetic beads were added to the plate and washed 3X using \SI{300}{\ul} of wash buffer. \SI{25}{\ul} of samples or standard were added to the plate and incubated for \SI{120}{\minute} at \SI{850}{\rpm} at \gls{rt} before washing analogously 3X with wash. \SI{12.5}{\ul} detection \glspl{mab} and \SI{25}{\ul} \gls{stppe} were sequentially added, incubated for \SI{30}{\minute} and vortexed, and washed analogously to the sample step. Finally, samples were resuspended in \SI{120}{\ul} reading buffer and analyzed via a BioRad Bioplex 200 plate reader. An 8 point log2 standard curve was used, and all samples were run with single replicates. Luminex data was preprocessed using R for inclusion in downstream analysis as follows. Any cytokine level that was over-range (`OOR >' in output spreadsheet) was set to the maximum value of the standard curve for that cytokine. Any value that was under-range (`OOR 90\% or noise-power) and lowest complexity, the top-performing models were identified. Driving variables, variable combinations, and model dimensionality tables were generated. The top-performing variable combinations were used to generate model ensembles. In this analysis, DataModeler’s SymbolicRegression function was used to develop explicit algebraic (linear and nonlinear) models. The fittest models were analyzed to identify the dominant variables using the VariablePresence function, the dominant variable combinations using the VariableCombinations function, and the model dimensionality (number of unique variables) using the ModelDimensionality function. CreateModelEnsemble was used to define trustable model ensembles using selected variable combinations and these were summarized (model expressions, model phenotype, model tree plot, ensemble quality, model quality, variable presence map, ANOVA tables, model prediction plot, exportable model forms) using the ModelSummaryTable function. Ensemble prediction and residual performance were respectively assessed via the EnsemblePredictionPlot and EnsembleResidualPlot subroutines. Model maxima (ModelMaximum function) and model minima (ModelMinimum function) were calculated and displayed using the ResponsePlotExplorer function. Trade-off performance of multiple responses was explored using the MultiTargetResponseExplorer and ResponseComparisonExplorer with additional insights derived from the ResponseContourPlotExplorer. Graphics and tables were generated by DataModeler. These model ensembles were used to identify predicted response values, potential optima in the responses, and regions of parameter values where the predictions diverge the most. Non-parametric tree-based ensembles were done through the randomForest, gbm, and cforest regression functions in R, for random forest, gradient boosted trees, and conditional inference forest models, respectively. Both random forest and conditional inference forest construct multiple decision trees in parallel, by randomly choosing a subset of features at each decision tree split, in the training stage. Random forest individual decision trees are split using the Gini Index, while conditional inference forest uses a statistical significance test procedure to select the variables at each split, reducing correlation bias. In contrast, gradient boosted trees construct regression trees in series through an iterative procedure that adapts over the training set. This model learns from the mistakes of previous regression trees in an iterative fashion to correct errors from its precursors’ trees (i.e. minimize mean squared errors). Prediction performance was evaluated using leave-one-out cross-validation (LOO)-R2 and permutation-based variable importance scores assessing \% increase of mean squared errors (MSE), relative influence based on the increase of prediction error, coefficient values for RF, GBM, and CID, respectively. Partial least squares regression was executed using the plsr function from the pls package in R while LASSO regression was performed using the cv.glmnet R package, both using leave-one-out cross-validation. Finally, the kernlab R package was used to construct the Support Vector Machine regression models. Parameter tuning was done for all models in a grid search manner using the train function from the caret R package using LOO-R2 as the optimization criteria. Specifically, the number of features randomly sampled as candidates at each split (mtry) and the number of trees to grow (ntree) were tuned parameters for random forest and conditional inference forest. In particular, minimum sum of weights in a node to be considered for splitting and the minimum sum of weights in a terminal node were manually tuned for building the CIF models. Moreover, GBM parameters such as the number of trees to grow, maximum depth of each tree, learning rate, and the minimal number of observations at the terminal node, were tuned for optimum LOO-R2 performance as well. For PLSR, the optimal number of components to be used in the model was assessed based on the standard error of the cross-validation residuals using the function selectNcomp from the pls package. Moreover, LASSO regression was performed using the cv.glmnet package with alpha = 1. The best lambda for each response was chosen using the minimum error criteria. Lastly, a fixed linear kernel (i.e. svmLinear) was used to build the SVM regression models evaluating the cost parameter value with best LOO-R2. Prediction performance was measured for all models using the final model with LOO-R2 tuned parameters. Table M2 shows the parameter values evaluated per model at the final stages of results reporting. \subsection{consensus analysis} Consensus analysis of the relevant variables extracted from each machine learning model was done to identify consistent predictive features of quality at the early stages of manufacturing. First importance scores for all features were measured across all ML models using varImp with caret R package except for scores for SVM which rminer R package was used. These importance scores were percent increase in mean squared error (MSE), relative importance through average increase in prediction error when a given predictor is permuted, permuted coefficients values, absolute coefficient values, weighted sum of absolute coefficients values, and relative importance from sensitivity analysis determined for RF, GBM, CIF, LASSO, PLSR, and SVM, respectively. Using these scores, key predictive variables were selected if their importance scores were within the 80th percentile ranking for the following ML methods: RF, GBM, CIF, LASSO, PLSR, SVM while for SR variables present in >30\% of the top-performing SR models from DataModeler (R2>= 90\%, Complexity >= 100) were chosen to investigate consensus except for NMR media models at day 4 which considered a combination of the top-performing results of models excluding lactate ppms, and included those variables which were in > 40\% of the best performing models. Only variables with those high percentile scoring values were evaluated in terms of their logical relation (intersection across ML models) and depicted using a Venn diagram from the venn R package. \section{results} \subsection{T cells can be grown on DMSs with lower IL2 concentrations} Prior to the main experiments in this aim, we performed a preliminary experiment to assess the effect of lowering the \gls{il2} concentration on the T cells grown with either bead or \gls{dms}. One of the hypotheses for the \gls{dms} system was that the higher cell density would enable more efficient cross-talk between T cells. Since \gls{il2} is secreted by activated T cells themselves, T cells in the \gls{dms} system may need less or no \gls{il2} if this hypothesis were true. % TODO this plots proportions look dumb % TODO explain what the NLS lines are in b % TODO plot the differences in lower IL2 concentrations to better show this \begin{figure*}[ht!] \begingroup \includegraphics{../figures/il2_modulation.png} \phantomsubcaption\label{fig:il2_mod_timecourse} \phantomsubcaption\label{fig:il2_mod_total} \phantomsubcaption\label{fig:il2_mod_mem} \phantomsubcaption\label{fig:il2_mod_flow} \endgroup \caption[T cells grown at varying IL2 concentrations] {\glspl{dms} grow T cells effectively at lower IL2 concentrations. \subcap{fig:il2_mod_timecourse}{Longitudinal cell counts of T cells grown with either bead or \glspl{dms} using varying IL2 concentrations} Day 14 counts of either \subcap{fig:il2_mod_total}{total cells} or \subcap{fig:il2_mod_mem}{\ptmem{} cells} plotted against \gls{il2} concentration. \subcap{fig:il2_mod_flow}{Flow cytometry plots of the \ptmem{} gated populations at day 14 of culture for each \gls{il2} concentration.} } \label{fig:il2_mod} \end{figure*} % TODO the nls stuff is a bit iffy We varied the concentration of \gls{il2} from \SIrange{0}{100}{\IU\per\ml} and expanded T cells as described in \cref{sec:tcellculture}. T cells grown with either method expanded robustly as \gls{il2} concentration was increased (\cref{fig:il2_mod_timecourse}). Surprisingly, neither the bead or the \gls{dms} group expanded at all with \SI{0}{\IU\per\ml} \gls{il2}. When examining the endpoint fold change after \SI{14}{\day}, we observe that the difference between the bead and \gls{dms} appears to be greater at lower \gls{il2} concentrations (\cref{fig:il2_mod_total}). This is further supported by fitting a non-linear least squares equation to the data following a hyperbolic curve (which should be a plausible model given that this curve describes receptor-ligand kinetics, which we can assume \gls{il2} to follow). Furthermore, the same trend can be seen when only examining the \ptmem{} cell expansion at day 14 (\cref{fig:il2_mod_mem}). In this case, the \ptmemp{} of the T cells seemed to be relatively close at higher \gls{il2} concentrations, but separated further at lower concentrations (\cref{fig:il2_mod_flow}) Taken together, these data do not support the hypothesis that the \gls{dms} system does not need \gls{il2} at all; however, it appears to have a modest advantage at lower \gls{il2} concentrations compared to beads. For this reason, we decided to investigate the lower range of \gls{il2} concentrations starting at \SI{10}{\IU\per\ml} throughout the remainder of this aim. % TODO this is not consistent with the next section since the responses are % different \subsection{DOE shows optimal conditions for expanded potent T cells} % TODO not all of these were actually use, explain why by either adding columns % or marking with an asterisk \begin{table}[!h] \centering \caption{DOE Runs} \label{tab:doe_runs} \input{../tables/doe_runs.tex} \end{table} % RESULT integrate this figure into the results paragraph \begin{figure*}[ht!] \begingroup \includegraphics{../figures/doe_responses_first.png} \phantomsubcaption\label{fig:doe_response_first_mem} \phantomsubcaption\label{fig:doe_response_first_cd4} \endgroup \caption[Response plots for first DOE] {Response plots from the first \gls{doe} experiment for \subcap{fig:doe_response_first_mem}{\ptmemp{}} and \subcap{fig:doe_response_first_cd4}{\pthp{}}. Each point is one run. } \label{fig:doe_response_first} \end{figure*} We conducted two consecutive \glspl{doe} to optimize the \pth{} and \ptmem{} responses for the \gls{dms} system. In the first \gls{doe} we, tested \pilII{} in the range of \SIrange{10}{30}{\IU\per\ml}, \pdms{} in the range of \SIrange{500}{2500}{\dms\per\ml}, and \pmab{} in the range of \SIrange{60}{100}{\percent}. % TODO explain why not all runs were used After performing the first \gls{doe} we augmented the original design matrix with an \gls{adoe} which was built with three goals in mind. Firstly we wished to validate the first \gls{doe} by assessing the strength and responses of each effect. Secondly, we wished to improve our confidence in regions that showed high complexity, such as the peak in the \gls{dms} concentration for the total \ptmem{} cell response. Thirdly, we wished to explore additional ranges of each response. Since \pilII{} and \pdms{} appeared to continue positively influence multiple responses beyond our tested range, we were curious if there was an optimum at some higher setting of either of these values. For this reason, we increased the \pilII{} to include \SI{40}{\IU\per\ml} and the \pdms{} to \SI{3500}{\dms\per\ml}. Note that it was impossible to go beyond \SI{100}{\percent} for the \pmab{}, so runs were positioned for this parameter with validation and confidence improvements in mind. The runs for each \gls{doe} were shown in \cref{tab:doe_runs}. \begin{figure*}[ht!] \begingroup \includegraphics{../figures/doe_responses.png} \phantomsubcaption\label{fig:doe_responses_mem} \phantomsubcaption\label{fig:doe_responses_cd4} \phantomsubcaption\label{fig:doe_responses_mem4} \phantomsubcaption\label{fig:doe_responses_ratio} \endgroup \caption[T cell optimization through Design of Experiments] {\gls{doe} methodology reveals optimal conditions for expanding T cell subsets. Responses vs IL2 concentration, \gls{dms} concentration, and functional \gls{mab} percentage are shown for \subcap{fig:doe_responses_mem}{total \ptmem{} T cells}, \subcap{fig:doe_responses_cd4}{total \pth{} T cells}, \subcap{fig:doe_responses_mem4}{total \ptmemh{} T cells}, and \subcap{fig:doe_responses_ratio}{ratio of CD4 and CD8 T cells in the \ptmem{} compartment}. Each point represents one run. } \label{fig:doe_responses} \end{figure*} \begin{table}[!h] \centering \caption{Total CD62L+CCR7+ T cell response (first order regression)} \label{tab:doe_mem1.tex} \input{../tables/doe_mem1.tex} \end{table} \begin{table}[!h] \centering \caption{Total CD62L+CCR7+ T cell response (third order regression)} \label{tab:doe_mem2.tex} \input{../tables/doe_mem2.tex} \end{table} \begin{table}[!h] \centering \caption{Total CD4+ T cell response} \label{tab:doe_cd4.tex} \input{../tables/doe_cd4.tex} \end{table} \begin{table}[!h] \centering \caption{Linear regression for total \ptmemh{} cells} \label{tab:doe_mem4.tex} \input{../tables/doe_mem4.tex} \end{table} \begin{table}[!h] \centering \caption{Linear regression for CD4:CD8 ratio in the \ptmem{} compartment} \label{tab:doe_ratio.tex} \input{../tables/doe_ratio.tex} \end{table} The response plots from both \glspl{doe} are shown in \cref{fig:doe_responses} for total \ptmem{} cells, total \pth{} cells, total \ptmemh{} cells, and CD4:CD8 ratio in the \ptmem{} compartment. In general, the responses for the first and second \gls{doe} seemed to overlap, although not perfectly. Interestingly, only the \ptmem{} response seemed to have anything more complex than a linear relationship, particularly in the case of \pilII{} and \pdms{}, which showed intermediate optimums (\cref{fig:doe_responses_mem}). In the case of \pilII{}, it was not clear if this optimum was simply due to a batch effect of being from the first or second \gls{doe}. The optimum for \pdms{} appeared in the same location albeit more pronounced in the second \gls{doe} so, giving more confidence to the location of this second order feature. The remainder of the responses showed mostly linear relationships in all parameter cases (\cref{fig:doe_responses_cd4,fig:doe_responses_mem4,fig:doe_responses_ratio}). % TODO it seems arbitrary that I went straight to a third order model, the real % reason is because it seemed weird that a second order model didn't find % anything to be significant We performed linear regression on the three input parameters as well as a binary parameter representing if a given run came from the first or second \gls{doe} (called `dataset'). Starting with the total \ptmem{} cells response, we fit a first order regression model using these four parameters (\cref{tab:doe_mem1.tex}). While \pilII{} was found to be a significant predictor, the model fit was extremely poor ($R^2$ of 0.331). This was not surprising given the apparent complexity of this response (\cref{fig:doe_responses_mem}). To obtain a better fit, we added second and third degree terms (\cref{tab:doe_mem2.tex}). Note that the dataset parameter was not included in the second order interaction as this was treated as a blocking variable, which are typically not assumed to have interaction effects. Also note that the response was log-transformed, which yielded a better fit. In this model many more parameters emerged as being significant, including the quadratic terms for \pdms{} and \pilII{}, in agreement with what can be qualitatively observed in the response plot (\cref{fig:doe_responses_mem}). Furthermore, the dataset parameter was weakly significant, indicating a possible batch effect between the \glspl{doe}. We should also note that despite many parameters being significant, this model was still only mediocre in describing this response; the $R^2$ was 0.741 but the adjusted $R^2$ was 0.583, indicating that our data might be underpowered for a model this complex. Further experiments beyond what was performed here may be needed to fully describe this response. % TODO combine these tables into one We performed linear regression on the other three responses, all of which performed much better than the \ptmem{} response as expected given the much lower apparent complexity in the response plots (\cref{fig:doe_responses_cd4,fig:doe_responses_mem4,fig:doe_responses_ratio}). All these models appeared to fit will, with $R^2$ and adjusted $R^2$ upward of 0.8. In all but the CD4:CD8 \ptmem{} ratio, the dataset parameter emerged as significant, indicating a batch effect between the \glspl{doe}. All other parameters except \pilII{} in the case of CD4:CD8 \ptmem{} ratio were significant predictors. \begin{figure*}[ht!] \begingroup \includegraphics{../figures/doe_sr_contour.png} \phantomsubcaption\label{fig:doe_sr_contour_mem4} \phantomsubcaption\label{fig:doe_sr_contour_ratio} \endgroup \caption[Contour plots for DOE responses] {Symbolic regression and contour plots reveal optimal conditions for \subcap{fig:doe_sr_contour_mem4}{\ptmemh{} cells} and \subcap{fig:doe_sr_contour_ratio}{CD4:CD8 ratio in the \ptmem{} compartment}. } \label{fig:doe_sr_contour} \end{figure*} We then visualized the total \ptmemh{} cells and CD4:CD8 \ptmem{} ratio using the response explorer in DataModeler to create contour plots around the maximum responses. For both, it appeared that maximizing all three input parameters resulted in the maximum value for either response (\cref{fig:doe_responses}). While not all combinations at and around this optimum were tested, the model nonetheless showed that there were no other optimal values or regions elsewhere in the model. % TODO this section header sucks \subsection{AI modeling reveals highly predictive species} \begin{figure*}[ht!] \begingroup \includegraphics{../figures/doe_luminex.png} \endgroup \caption[Cytokine release profile of T cells from DOE] {T cells show robust and varying cytokine responses over time} \label{fig:doe_luminex} \end{figure*} Due to the heterogeneity of the multivariate data collected and knowing that no single model structure is perfect for all applications, we implemented an agnostic modeling approach to better understand these TN+TCM responses. To achieve this, a consensus analysis using seven machine learning (ML) techniques, Random Forest (RF), Gradient Boosted Machine (GBM), Conditional Inference Forest (CIF), Least Absolute Shrinkage and Selection Operator (LASSO), Partial Least-Squares Regression (PLSR), Support Vector Machine (SVM), and DataModeler’s Symbolic Regression (SR), was implemented to molecularly characterize TN+TCM cells and to extract predictive features of quality early on their expansion process (Fig.1d-e). % TODO this table looks like crap, break it up into smaller tables \begin{table}[!h] \centering \caption{Results for data-driven modeling} \label{tab:mod_results} \input{../tables/model_results.tex} \end{table} SR models achieved the highest predictive performance (R2>93\%) when using multi-omics predictors for all endpoint responses (\cref{tab:mod_results}). SR achieved R2>98\% while GBM tree-based ensembles showed leave-one-out cross-validated R2 (LOO-R2) >95\% for CD4+ and CD4+/CD8+ TN+TCM responses. Similarly, LASSO, PLSR, and SVM methods showed consistent high LOO-R2, 92.9\%, 99.7\%, and 90.5\%, respectively, to predict the CD4+/CD8+ TN+TCM. Yet, about 10\% reduction in LOO-R2, 72.5\%-81.7\%, was observed for CD4+ TN+TCM with these three methods. Lastly, SR and PLSR achieved R2>90\% while other ML methods exhibited exceedingly variable LOO-R2 (0.3\%,RF-51.5\%,LASSO) for CD8+ TN+TCM cells. \begin{figure*}[ht!] \begingroup \includegraphics{../figures/sr_omics.png} \endgroup \caption[Symbolic Regression Cytokine Dependencies] {Multi-omics culturing media prediction profiles at day 6 using symbolic regression.} \label{fig:sr_omics} \end{figure*} The top-performing technique, SR, showed that the median aggregated predictions for CD4+ and CD8+ TN+TCM cells increases when IL2 concentration, IL15, and IL2R increase while IL17a decreases in conjunction with other features. These patterns combined with low values of DMS concentration and GM-CSF uniquely characterized maximum CD8+ TN+TCM. Meanwhile, higher glycine but lower IL13 in combination with others showed maximum CD4+ TN+TCM predictions (\cref{fig:sr_omics}). \begin{figure*}[ht!] \begingroup \includegraphics{../figures/modeling_flower.png} \phantomsubcaption\label{fig:mod_flower_48r} \phantomsubcaption\label{fig:mod_flower_cd4} \endgroup \caption[Data-Driven \gls{cqa} identification] {Data-driven modeling using techniques with regularization reveals species predictive species which are candidates for \glspl{cqa}. Flower plots are shown for \subcap{fig:mod_flower_48r}{CD4:CD8 ratio} and \subcap{fig:mod_flower_cd4}{total \ptmemh{} cells}. The left and right columns includes models that were trained only on the secretome and metabolome respectively. Each flower on each plot represents one model, moving toward the center indicates higher agreement between models.} \label{fig:mod_flower} \end{figure*} Selecting CPPs and CQAs candidates consistently for T cell memory is desired. Here, \gls{tnfa} was found in consensus across all seven ML methods for predicting CD4+/CD8+ TN+TCM when considering features with the highest importance scores across models (Fig.3a;Methods). Other features, IL2R, IL4, IL17a, and DMS concentration, were commonly selected in >=5 ML methods (Fig.3a,c). Moreover, IL13 and IL15 were found predictive in combination with these using SR (Supp.Table.S4). \begin{figure*}[ht!] \begingroup \includegraphics{../figures/nmr_cors.png} \phantomsubcaption\label{fig:nmr_cors_lactate} \phantomsubcaption\label{fig:nmr_cors_formate} \phantomsubcaption\label{fig:nmr_cors_glucose} \phantomsubcaption\label{fig:nmr_cors_matrix} \endgroup \caption[NMR Day 4 correlations] {\gls{nmr} features at day 4 are strongly correlated with each other and the response variables. Highly correlated relationships are shown for \subcap{fig:nmr_cors_lactate}{lactate}, \subcap{fig:nmr_cors_formate}{formate}, and \subcap{fig:nmr_cors_glucose}{glucose}. Blue and blue connections indicate positive and negative correlations respectively. The threshold for visualizing connections in all cases was 0.8. \subcap{fig:nmr_cors_matrix}{The correlation matrix for all predictive features and the total \ptmemh{} response.} } \label{fig:nmr_cors} \end{figure*} \section{discussion} % optimization of process features % TODO this sounds like total fluff CPPs modeling and understanding are critical to new product development and in cell therapy development, it can have life-saving implications. The challenges for effective modeling grow with the increasing complexity of processes due to high dimensionality, and the potential for process interactions and nonlinear relationships. Another critical challenge is the limited amount of available data, mostly small DOE datasets. SR has the necessary capabilities to resolve the issues of process effects modeling and has been applied across multiple industries12. SR discovers mathematical expressions that fit a given sample and differs from conventional regression techniques in that a model structure is not defined a priori13. Hence, a key advantage of this methodology is that transparent, human-interpretable models can be generated from small and large datasets with no prior assumptions14,15. Since the model search process lets the data determine the model, diverse and competitive (e.g., accuracy, complexity) model structures are typically discovered. An ensemble of diverse models can be formed where its constituent models will tend to agree when constrained by observed data yet diverge in new regions. Collecting data in these regions helps to ensure that the target system is accurately modeled, and its optimum is accurately located14,15. Exploiting these features allows adaptive data collection and interactive modeling. Consequently, this adaptive-DOE approach is useful in a variety of scenarios, including maximizing model validity for model-based decision making, optimizing processing parameters to maximize target yields, and developing emulators for online optimization and human understanding14,15. % predictive features An in-depth characterization of potential DMS-based T-cell CQAs includes a list of cytokine and NMR features from media samples that are crucial in many aspects of T cell fate decisions and effector functions of immune cells. Cytokine features were observed to slightly improve prediction and dominated the ranking of important features and variable combinations when modeling together with NMR media analysis and process parameters (Fig.3b,d). Predictive cytokine features such as \gls{tnfa}, IL2R, IL4, IL17a, IL13, and IL15 were biologically assessed in terms of their known functions and activities associated with T cells. T helper cells secrete more cytokines than T cytotoxic cells, as per their main functions, and activated T cells secrete more cytokines than resting T cells. It is possible that some cytokines simply reflect the CD4+/CD8+ ratio and the activation degree by proxy proliferation. However, the exact ratio of expected cytokine abundance is less clear and depends on the subtypes present, and thus examination of each relevant cytokine is needed. IL2R is secreted by activated T cells and binds to IL2, acting as a sink to dampen its effect on T cells16. Since IL2R was much greater than IL2 in solution, this might reduce the overall effect of IL2, which could be further investigated by blocking IL2R with an antibody. In T cells, TNF can increase IL2R, proliferation, and cytokine production18. It may also induce apoptosis depending on concentration and alter the CD4+ to CD8+ ratio17. Given that TNF has both a soluble and membrane-bound form, this may either increase or decrease CD4+ ratio and/or memory T cells depending on the ratio of the membrane to soluble TNF18. Since only soluble TNF was measured, membrane TNF is needed to understand its impact on both CD4+ ratio and memory T cells. Furthermore, IL13 is known to be critical for Th2 response and therefore could be secreted if there are significant Th2 T cells already present in the starting population19. This cytokine has limited signaling in T cells and is thought to be more of an effector than a differentiation cytokine20. It might be emerging as relevant due to an initially large number of Th2 cells or because Th2 cells were preferentially expanded; indeed, IL4, also found important, is the conical cytokine that induces Th2 cell differentiation (Fig.3). The role of these cytokines could be investigated by quantifying the Th1/2/17 subsets both in the starting population and longitudinally. Similar to IL13, IL17 is an effector cytokine produced by Th17 cells21 thus may reflect the number of Th17 subset of T cells. GM-CSF has been linked with activated T cells, specifically Th17 cells, but it is not clear if this cytokine is inducing differential expansion of CD8+ T cells or if it is simply a covariate with another cytokine inducing this expansion22. Finally, IL15 has been shown to be essential for memory signaling and effective in skewing CAR-T cells toward the Tscm phenotype when using membrane-bound IL15Ra and IL15R23. Its high predictive behavior goes with its ability to induce large numbers of memory T cells by functioning in an autocrine/paracrine manner and could be explored by blocking either the cytokine or its receptor. Moreover, many predictive metabolites found here are consistent with metabolic activity associated with T cell activation and differentiation, yet it is not clear how the various combinations of metabolites relate with each other in a heterogeneous cell population. Formate and lactate were found to be highly predictive and observed to positively correlate with higher values of total live CD4+ TN+TCM cells (Fig.5a-b;Supp.Fig.28-S30,S38). Formate is a byproduct of the one-carbon cycle implicated in promoting T cell activation24. Importantly, this cycle occurs between the cytosol and mitochondria of cells and formate excreted25. Mitochondrial biogenesis and function are shown necessary for memory cell persistence26,27. Therefore, increased formate in media could be an indicator of one-carbon metabolism and mitochondrial activity in the culture. In addition to formate, lactate was found as a putative CQA of TN+TCM. Lactate is the end-product of aerobic glycolysis, characteristic of highly proliferating cells and activated T cells28,29. Glucose import and glycolytic genes are immediately upregulated in response to T cell stimulation, and thus generation of lactate. At earlier time-points, this abundance suggests a more robust induction of glycolysis and higher overall T cell proliferation. Interestingly, our models indicate that higher lactate predicts higher CD4+, both in total and in proportion to CD8+, seemingly contrary to previous studies showing that CD8+ T cells rely more on glycolysis for proliferation following activation30. It may be that glycolytic cells dominate in the culture at the early time points used for prediction, and higher lactate reflects more cells. % TODO not sure how much I should include here since I didn't do this analysis % AT ALL % Ethanol patterns are difficult to interpret since its production in mammalian % cells is still poorly understood31. Fresh media analysis indicates ethanol % presence in the media used, possibly utilized as a carrier solvent for certain % formula components. However, this does not explain the high variability and % trend of ethanol abundance across time (Supp.Fig.S25-S27). As a volatile % chemical, variation could be introduced by sample handling throughout the % analysis process. Nonetheless, it is also possible that ethanol excreted into % media over time, impacting processes regulating redox and reactive oxygen % species which have previously been shown to be crucial in T cell signaling and % differentiation32. % this looks fine since it is just parroting sources, just need to paraphrase a % little Metabolites that consistently decreased over time are consistent with the primary carbon source (glucose) and essential amino acids (BCAA, histidine) that must be continually consumed by proliferating cells. Moreover, the inclusion of glutamine in our predictive models also suggests the importance of other carbon sources for certain T cell subpopulations. Glutamine can be used for oxidative energy metabolism in T cells without the need for glycolysis30. Overall, these results are consistent with existing literature that show different T cell subtypes require different relative levels of glycolytic and oxidative energy metabolism to sustain the biosynthetic and signaling needs of their respective phenotypes33,34. It is worth noting that the trends of metabolite abundance here are potentially confounded by the partial replacement of media that occurred periodically during expansion (Methods), thus likely diluting some metabolic byproducts (i.e. formate, lactate) and elevating depleted precursors (i.e. glucose, amino acids). More definitive conclusions of metabolic activity across the expanding cell population can be addressed by a closed system, ideally with on-line process sensors and controls for formate, lactate, along with ethanol and glucose. \chapter{aim 2b}\label{aim2b} \section{introduction} \section{methods} \subsection{DMSs temporal modulation} % TODO The concentration for the surface marker cleavage experiment was much % higher, if that matters \glspl{dms} were digested in active T cell cultures via addition of sterile \product{\gls{colb}}{Sigma}{11088807001} or \product{\gls{cold}}{Sigma}{11088858001}. Collagenase was dissolved in \product{\gls{hbss}}{Gibco}{14025-076} or \product{TexMACS}{\miltenyi}{170-076-307} at approximately \SI{100}{\ug\per\ml}. This solution was added to T cell cultures at a 1:1 ratio in place of plain media normally used to feed the cells during the regular media addition cycle at day 4. Cultures were then incubated as described in \cref{sec:tcellculture}, and the \glspl{dms} were verified to have been digested after \SI{24}{\hour}. Adding \gls{dms} was relatively much simpler; the number of \gls{dms} used per area on day 0 was scaled up by 3 on day 4 to match the change from a 96 well plate to a 24 well plate, effectively producing a constant activation signal. \subsection{mass cytometry and clustering analysis} T cells were stained using a \product{34 \gls{cytof} marker panel}{Fluidigm}{201322} and \product{cisplatin}{Fluidigm}{201064} which were used according to the manufacturer’s instructions. \numrange{2e6}{3e6} stained cells per group were analyzed on a Fluidigm Helios. Unbiased cell clusters were obtained using \gls{spade} analysis by pooling three representative \gls{fcs} files and running the \gls{spade} pipeline with k-means clustering (k = 100), arcsinh transformation with cofactor 5, density calculation neighborhood size of 5 and local density approximation factor of 1.5, target density of 20000 cells, and outlier density cutoff of \SI{1}{\percent}. All markers in the \gls{cytof} panel were used in the analysis \subsection{integrin blocking experiments} To block \gls{a2b1} and \gls{a2b2}, active T cell cultures with \gls{dms} were supplemented with \product{\anti{\gls{a2b1}}}{Sigma}{MAB1973Z} and \product{\anti{\gls{a2b2}}}{Sigma}{MAB1950Z} (both \gls{leaf}) at indicated concentrations and timepoints. T cells were grown as described in \cref{sec:tcellculture}. \gls{a2b1} and \gls{a2b2} were verified to be present on active T cell cultures by staining with \product{\anti{\gls{a2b1}}-\gls{apc}}{\bl}{328313} and \product{\anti{\gls{a2b2}}-\gls{fitc}}{\bl}{359305} on day 6 of culture and analyzing via a BD Accuri flow cytometer. \subsection{IL15 blocking experiments} To block the \gls{il15r}, we supplemented T cell cultures activated with \gls{dms} with either \product{\anti{\gls{il15r}}}{Rnd}{AF247} or \product{\gls{igg} isotype control}{RnD}{AB-108-C} at the indicated timepoints and concentrations. T cells were grown as otherwise described in \cref{sec:tcellculture} with the exception that volumes were split by $\frac{1}{3}$ to keep the culture volume constant and minimize the amount of \gls{mab} required. To block soluble \gls{il15}, we supplemented analogously with \product{\anti{\gls{il15}}}{RnD}{EEP0419081} or \product{\gls{igg} isotype control}{\bl}{B236633}. \section{results} \subsection{adding or removing DMSs alters expansion and phenotype} % TODO state what collagenase actually targets We hypothesized that adding or removing \gls{dms} in the middle of an active culture would alter the activation signal and hence the growth trajectory and phenotype of T cells. While adding \glspl{dms} was simple, the easiest way to remove \glspl{dms} was to use enzymatic digestion. Collagenase is an enzyme that specifically targets the blabla domain on collagen. Since our \glspl{dms} are composed of porcine-derived collagen, this enzyme should target the \gls{dms} while sparing the cells. We tested this specific hypothesis using either \gls{colb}, \gls{cold} or \gls{hbss}, and stained the cells using a typical marker panel to assess if any of the markers were cleaved off by the enzyme which would bias our final readout. We observed that the marker histograms in the \gls{cold} group were similar to that of the buffer group, while the \gls{colb} group visibly lowered CD62L and CD4, indicating partial enzymatic cleavage (\cref{fig:collagenase_fx}). Based on this result, we used \gls{cold} moving forward. % TODO this figure is tall and skinny like me \begin{figure*}[ht!] \begingroup \includegraphics{../figures/collagenase.png} \endgroup \caption[Effects Collagenase Treatment on T cells] {T cells treated with either \gls{colb}, \gls{cold}, or buffer and then stained for various surface markers and analyzing via flow cytometry.} \label{fig:collagenase_fx} \end{figure*} When either adding more \glspl{dms}, removing \glspl{dms} using \gls{cold}, or doing nothing, we observed that, counterintuitively, cell growth seemed to be inhibited in the \textit{added} group while the cells seemed to grow faster in the \textit{removed} group relative to the \textit{no change} group (\cref{fig:add_rem_growth}). Additionally, the \textit{removed} group seemed to have a negative growth rate in the final \SI{4}{\day} of culture, indicating that either the lack activation signal had slowed the cell growth down or that the cells were growing fast enough to outpace the media feeding schedule. The viability was the same between all groups, indicating that this negative growth rate and the lower growth rate in the \textit{added} group were likely not due to cell death (\cref{fig:add_rem_viability}). Interestingly, the \textit{added} group had significantly higher \pth{} cells compared to the \textit{no change} group, and the inverse was true for the \textit{removed} group (\cref{fig:add_rem_cd4}). These results show that the growth rate and phenotype are fundamentally altered by changing the number of \glspl{dms} temporally. % TODO this figure still says "carrier" \begin{figure*}[ht!] \begingroup \includegraphics{../figures/add_remove_endpoint.png} \phantomsubcaption\label{fig:add_rem_growth} \phantomsubcaption\label{fig:add_rem_viability} \phantomsubcaption\label{fig:add_rem_cd4} \endgroup \caption[Endpoint results from adding/removing \gls{dms} on day 4] {Changing \gls{dms} concentration on day 4 has profound effects on phenotype and growth. \subcap{fig:add_rem_growth}{Longitudinal fold change}, \subcap{fig:add_rem_viability}{longitudinal viability}, and \subcap{fig:add_rem_cd4}{day 14 \pthp{}} of T cell cultures with \glspl{dms} added, removed, or kept the same on day 4. } \label{fig:add_rem} \end{figure*} \begin{figure*}[ht!] \begingroup \includegraphics{../figures/spade_gates.png} \endgroup \caption[SPADE Gating Strategy] {Gating strategy for quantifying early-differentiated T cells via \gls{spade}.} \label{fig:spade_gates} \end{figure*} % TODO this needs some better annotations % TODO put the quant graph before the tsne stuff \begin{figure*}[ht!] \begingroup \includegraphics{../figures/add_remove_spade.png} \phantomsubcaption\label{fig:spade_msts} \phantomsubcaption\label{fig:spade_tsne_all} \phantomsubcaption\label{fig:spade_tsne_stem} \phantomsubcaption\label{fig:spade_quant} \endgroup \caption[SPADE and tSNE analysis temporally-modified DMS concentration] {Removing \glspl{dms} leads to a higher fraction of potent stem-memory T cells compared to both adding and not changing the \gls{dms} concentration at day 4. \subcap{fig:spade_msts}{SPADE plots of CD4, CD45RA, CD27, and CD45RO expression on T cells. All cells from the added, removed, or no change groups were pooled and clustered at once.} \subcap{fig:spade_tsne_all}{\gls{tsne} plots of all cells pooled from all groups.} \subcap{fig:spade_tsne_stem}{\gls{tsne} plots of T cells from all groups manually gated on \cdp{8}\cdp{27}\cdp{45RO}.} \subcap{fig:spade_quant}{T cells from SPADE plots clustered by expression in (\subref{fig:spade_msts}) quantified against total cell number from each group.} } \label{fig:spade} \end{figure*} We next asked what the effect of removing the \glspl{dms} would have on other phenotypes, specifically \gls{tcm} and \gls{tscm} cells. To this end we stained cells using a 34-marker mass cytometry panel and analyzed them using a Fluidigm Helios. After pooling the \gls{fcs} file events from each group and analyzing them via \gls{spade} we see that there is a strong bifurcation of CD4 and CD8 T cells. We also observe that among CD27, CD45RA, and CD45RO (markers commonly used to identify \gls{tcm} and \gls{tscm} subtypes) we see clear `metaclusters' composed of individual \gls{spade} clusters which are high for that marker (\cref{fig:spade_msts,fig:spade_gates}). We then gated each of these metaclusters according to their marker levels and assigned them to one of three phenotypes for both the CD4 and CD8 compartments: \gls{tcm} (high CD45RO, low CD45RA, high CD27), \gls{tscm} (low CD45RO, high CD45RA, high CD27), and `transitory' \gls{tscm} cells (mid CD45RO, mid CD45RA, high CD27). Together these represent low differentiated cells which should be highly potent as anti-tumor therapies. When quantifying the number of cells from each experimental group in these phenotypes, we clearly see that the number of lower differentiated cells is much higher in the \textit{no change} or \textit{removed} groups compared to the \textit{added} group (\cref{fig:spade_quant}). Furthermore, the \textit{removed} group had a much higher fraction of \gls{tscm} cells compared to the \textit{no change} group, which had more `transitory \gls{tscm} cells'. The majority of these cells were \cdp{8} cells. When analyzing the same data using \gls{tsne}, we observe a higher fraction of CD27 and lower fraction of CD45RO in the the \textit{removed} group (\cref{fig:spade_tsne_all}). When manually gating on the CD27+CD45RO- population, we see there is higher density in the \textit{removed} group, indicating more of this population (\cref{fig:spade_tsne_stem}). Together, these data indicate that removing \glspl{dms} at lower timepoints leads to potentially higher expansion, lower \pthp{}, and higher fraction of lower differentiated T cells such as \gls{tscm}, and adding \gls{dms} seems to do the inverse. \subsection{blocking integrin binding does not alter expansion or phenotype} % BACKGROUND add background into why integrins are important One of the reasons the \gls{dms} platform might be performing better than the beads is the fact that they are composed of gelatin, which is a collagen derivative. The beads are simply \gls{mab} attached to a polymer resin coated onto an iron oxide core, and thus have no analogue for collagen. Collagen domains present on the \gls{dms} group could be creating pro-survival and pro-expansion signals to the T cells through \gls{a2b1} and \gls{a2b2}, causing them to grow better in the \gls{dms} system. % TODO perhaps these figs should be combined % TODO actually make the captions for these % TODO add some background into why integrins are important and the proposed mechanism % TODO add an experimental timeline to these showing when I added the mabs \begin{figure*}[ht!] \begingroup \includegraphics{../figures/integrin_1.png} \phantomsubcaption\label{fig:inegrin_1_fc} \phantomsubcaption\label{fig:inegrin_1_mem} \phantomsubcaption\label{fig:inegrin_1_cd49} \endgroup \caption[Integrin blocking I] {Blocking with integrin does not lead to differences in memory or growth. } \label{fig:integrin_1} \end{figure*} % TABLE add regression table showing the results from this analysis We tested this hypothesis by adding blocking \glspl{mab} against \gls{a2b1} and/or \gls{a2b2} to running T cell cultures activated using the \glspl{dms}. These block \glspl{mab} were added at day 6 of culture when \gls{a2b1} and \gls{a2b2} were known to be expressed {\#}. We found that the fold expansion was identical in all the blocked groups vs the unblocked control group (\cref{fig:inegrin_1_fc}). Furthermore, we observed that the \ptmemp{} (total and across the CD4/CD8 compartments) was not significantly different between any of the groups (\cref{fig:inegrin_1_mem}). We also noted that \gls{a2b1} and \gls{a2b2} were present on the surface of a significant subset of T cells at day 6, showing that the target we wished to block was present (\cref{fig:inegrin_1_cd49}). \begin{figure*}[ht!] \begingroup \includegraphics{../figures/integrin_2.png} \phantomsubcaption\label{fig:inegrin_2_fc} \phantomsubcaption\label{fig:inegrin_2_mem} \endgroup \caption[Integrin blocking II] {Blocking with integrin does not lead to differences in memory or growth. } \label{fig:integrin_2} \end{figure*} Since this last experiment gave a negative result, we decided to hit \gls{a2b1} and \gls{a2b2} harder by adding blocking \glspl{mab} at more timepoints between day 0 and day 6, hypothesizing that the majority of the signaling would be during the period of culture where the \gls{dms} surface concentration was at its maximum. Once again, we observed no difference between any of the blocked conditions and the unblocked controls in regard to expansion (\cref{fig:inegrin_2_fc}). Furthermore, none of the \ptmemp{} readouts (total, CD4, or CD8) were statistically different between groups (\cref{fig:inegrin_2_mem}). Taken together, these data suggest that the advantage of the \gls{dms} platform is not due to signaling through \gls{a2b1} or \gls{a2b2}. \subsection{blocking IL15 signaling does not alter expansion or phenotype} % BACKGROUND why is IL15 important? % TODO cite the luminex data \gls{il15} is a cytokine responsible for memory T cell survival and maintenance. Furthermore, we observed in other experiments that it is secreted to a much greater extend in \gls{dms} compared to bead cultures. One of our driving hypotheses in designing the \gls{dms} system was that the higher cell density would lead to greater local signaling. Since we observed higher \ptmemp{} across many conditions, we hypothesized that \gls{il15} may be responsible for this, and further that the unique \textit{cis/trans} activity of \gls{il15} may be more active in the \gls{dms} system due to higher cell density. % TODO actually add captions % TODO add fold change and viability to these % TODO maybe combine these % TODO add some background into why IL15 is important and the proposed mechanism \begin{figure*}[ht!] \begingroup \includegraphics{../figures/il15_blockade_1.png} \phantomsubcaption\label{fig:il15_1_fc} \phantomsubcaption\label{fig:il15_1_viability} \phantomsubcaption\label{fig:il15_1_mem} \endgroup \caption[IL15 blocking I] {Blocking with IL15 does not lead to differences in memory or growth. } \label{fig:il15_1} \end{figure*} % TODO how did I determine how much to add? % TODO how often was this added? % TODO just gate these as normal because this looks sketchy We first tested this hypothesis by blocking \gls{il15r} with either a specific \gls{mab} or an \gls{igg} isotype control. We observed no difference in the expansion rate of blocked or unblocked cells (this experiment also had bead-based groups but they did not expand well and thus were not included) (\cref{fig:il15_1_fc}). Furthermore, there were no differences in viability between any group (\cref{fig:il15_1_viability}). We also performed flow cytometry to asses the \ptmemp{} and \pthp{} outputs. Without even gating the samples, simply lining up their histograms showed no difference between any of the markers, and by extension showing no difference in phenotype (\cref{fig:il15_1_mem}). \begin{figure*}[ht!] \begingroup \includegraphics{../figures/il15_blockade_2.png} \phantomsubcaption\label{fig:il15_2_fc} \phantomsubcaption\label{fig:il15_2_viability} \phantomsubcaption\label{fig:il15_2_mem} \endgroup \caption[IL15 blocking II] {Blocking with IL15 does not lead to differences in memory or growth. } \label{fig:il15_2} \end{figure*} We next tried blocking soluble \gls{il15} itself using either a \gls{mab} or an \gls{igg} isotype control. Similarly, we observed no difference between fold change, viability, or marker histograms between any of these markers, showing that blocking \gls{il15} led to no difference in growth or phenotype. % TODO this can probably be worded more specifically in terms of the cis/trans % action of IL15 In summary, this data did not support the hypothesis that the \gls{dms} platform gains its advantages via the \gls{il15} pathway. \section{discussion} This work provides insight for how the \gls{dms} operates and may be optimized further. The data showing increased \pthp{} when \glspl{dms} are added and the reverse when removed is consistent with other data we produced via \gls{doe} showing that higher \gls{dms} concentrations lead to higher \pthp{} (\cref{fig:doe_responses_cd4,fig:add_rem_cd4}). The difference in this case is that we showed that altering activation signal analogously affects the \pthp{} in the dimension of time as well as space. A similar trend was observed with memory T cells in this aim. Our previous \gls{doe} data showed that, to a point, lower \gls{dms} concentration leads to higher \ptmemp{} (\cref{fig:doe_responses_mem}). In this aim, we showed that decreasing activation signal temporally by removing \glspl{dms} leads to the same effect in the \gls{tcm}, \gls{tscm} and `transitory' \gls{tscm} populations, (all of which are included in the \ptmem{} phenotype). Taken together, these imply that temporally or spatially altering the \gls{dms} concentration, and thus the activation signal, has similar effects. % TODO this sounds like background? % There are several plausible explanations for the observed phenotypic differences % between beads and DMSs. First, the DMSs are composed of a collagen derivative % (gelatin); collagen has been shown to costimulate activated T cells via % \gls{a2b1} and \gls{a2b2}, leading to enhanced proliferation, increased % \gls{ifng} production, and upregulated CD25 (IL2R$\upalpha$) surface % expression8,10,11,41,42. While we did not find support for our hypothesis that the \gls{dms} signal through the \gls{a2b1} and/or \gls{a2b2} receptors, we can speculate as to why either this experiment failed and may be done better in the future, or why these receptors may simply be irrelevant for our system. On the first point, we did not verify that these \glspl{mab} indeed blocked the receptor we were targeting. There has been evidence from other groups that these particular clones work at the concentrations we used {\#}. This does not necessarily mean that the \glspl{mab} we obtained were functional in blocking their intended targets (although they were from a reputable manufacturer, \bl). Furthermore, we can safely rule out the possibility that the \glspl{mab} never reached their targets, as they were added immediately after the T cells were resuspended as required for cell counting, hence their resting clustered state was disrupted. % TODO define Bite On the second point, the collagen domains may not even be relevant to our system depending on the nature of the \gls{stp} coating. We intended by design for the system to be fully coated or nearly fully-coated with \gls{stp} (\cref{fig:stp_coating}). Thus the domains that \gls{a2b1} and \gls{a2b2} may be targeting could be sterically hindered by a layer of \gls{stp}, and if not that, also a layer of CD3/CD28 \glspl{mab}. The other possibility is that these domains are simply denatured to beyond recognition due to the fabrication process for the microcarriers we used (which involves a proprietary cross-linking step to make the material autoclave-safe). Either of these could be tested and verified by staining the \glspl{dms} with a fluorescently-tagged \gls{mab} (or something smaller such as a BiTE to reduce the possibility of steric hindrance) and verifying binding via confocal microscopy or indirect protein quantification as we do for the \gls{qc} of the \gls{dms}. If this test came back negative, we would be fairly confident that the \gls{a2b1} and \gls{a2b1} domains are either unreachable or unrecognizable. % DISCUSSION not sure exactly how to explain this We also failed to uphold our hypothesis that the \gls{dms} system gains its advantage via \gls{il15} signaling. % TODO not sure if this belongs here, although it might make sense to offer % alternative explanations of why the DMSs "work" given this negative data % Second, there is evidence that providing a larger % contact area for T cell activation provides greater stimulation16,43; the DMSs % have a rougher interface than the 5 µm magnetic beads, and thus could facilitate % these larger contact areas. Third, the DMSs may allow the T cells to cluster % more densely compared to beads, as evidenced by the large clusters on the % outside of the DMSs (Figure 1f) as well as the significant fraction of DMSs % found within their interiors (Supplemental Figure 2a and b). This may alter the % local cytokine environment and trigger different signaling pathways. % Particularly, IL15 and IL21 are secreted by T cells and known to drive memory % phenotype44–46. We noted that the IL15 and IL21 concentration was higher in a % majority of samples when comparing beads and DMSs across multiple timepoints % (Supplemental Figure 18) in addition to many other cytokines. IL15 and IL21 are % added exogenously to T cell cultures to enhance memory frequency,45,47 and our % data here suggest that the DMSs are better at naturally producing these % cytokines and limiting this need. Furthermore, IL15 unique signals in a trans % manner in which IL15 is presented on IL15R to neighboring cells48. The higher % cell density in the DMS cultures would lead to more of these trans interactions, % and therefore upregulate the IL15 pathway and lead to more memory T cells. \chapter{aim 3}\label{aim3} \section{introduction} \section{methods} \subsection{CD19-CAR T cell generation} % METHOD describe how T cells were grown for this aim % METHOD describe how the luciferase cells were generated (eg the kwong lab) \subsection{\invivo{} therapeutic efficacy in NSG mice model} % TODO use actual product numbers for mice All mice in this study were male \gls{nsg} mice from Jackson Laboratories. At day 0 (-7 day relative to T cell injection), 1e6 firefly luciferase-expressing \product{Nalm-6 cells}{ATCC}{CRL-3273} suspended in ice-cold PBS were injected via tail vein into each mouse. At day 7, saline or CAR T cells at the indicated doses from either bead or DMS-expanded T cell cultures (for 14 days) were injected into each mouse via tail vein. Tumor burden was quantified longitudinally via an \gls{ivis} Spectrum (Perkin Elmer). Briefly, 200ug/mice luciferin at 15 mg/ml in PBS was injected intraperitoneally under isoflurane anesthesia into each mouse and waited for at least 10 minutes before imaging. Mice were anesthetized again and imaged using the \gls{ivis}. Mice from each treatment group/dose were anesthetized, injected, and imaged together, and exposure time of the \gls{ivis} was limited to avoid saturation based on the signal from the saline group. \gls{ivis} images were processed by normalizing them to common minimum and maximum photon counts and total flux was estimated in terms of photons/second. Endpoint for each mouse was determined by \gls{iacuc} euthanasia criteria (hunched back, paralysis, blindness, lethargy, and weight loss). Mice were euthanized according to these endpoint criteria using carbon dioxide asphyxiation. \subsection{statistics} For the \invivo{} model, the survival curves were created and statistically analyzed using GraphPad Prism using the Mantel-Cox test to assess significance between survival groups. \section{results} We asked if the higher memory/naive phenotype and more balanced CD4/CD8 ratio of our \gls{dms}-expanded CAR T cells would lead to better anti-tumor potency in vivo compared to bead-expanded CAR T cells. We also asked if this superior anti-tumor potency would hold true at lower doses of CAR expressing T cells in the DMS group vs the bead group. To test this, we used a human xenograft model of B cell \gls{all} by intravenously injecting \gls{nsg} mice with \num{1e6} Nalm-6 tumor cells expression firefly luciferase\cite{Fraietta2018}. After \SI{7}{\day} of tumor cell growth (\cref{fig:mouse_dosing_overview}), we intravenously injected saline or three doses (high, medium, and low) of \gls{dms} T cells from either bead or DMS cultures expanded for \SI{14}{\day}. We quantified total \gls{dms} expressing T cell percentage for bead and \gls{dms} groups using the \gls{ptnl} assay (\cref{tab:mouse_dosing_results}). Before injecting the T cells into the mice, we quantified their phenotype and growth. We observed that for this expansion, the bead and \gls{dms} T cells produced similar numbers of \ptmem{} T cells, and the beads even had a higher fraction of CD45RA, which is present on lower-differentiated naive and stem-memory T cells (\cref{fig:mouse_dosing_qc_mem}). However, the \pthp{} of the final product was higher in \gls{dms} (\cref{fig:mouse_dosing_qc_cd4}). The \gls{dms} T cells also expanded more robustly than the beads (\cref{fig:mouse_dosing_qc_growth}). In the Nalm-6/\gls{nsg} xenograft model, we observed lower tumor burden and significantly longer survival of bead and \gls{dms}-treated mice at all doses compared to the saline groups (\cref{fig:mouse_dosing_ivis}). Importantly, at each dose we observed that the \gls{dms}-treated mice had much lower tumor burden and significantly higher survival than their bead-treated counterparts (\cref{fig:mouse_dosing_ivis_survival}). When factoring the percentage T cells in each dose that expressed the \gls{car}, we note that survival of the low \gls{dms} dose (which had similar total \gls{car} T cells compared to the bead medium dose and less than the bead high dose) is significantly higher than that of both the bead medium dose and the bead high dose (\cref{fig:mouse_dosing_ivis_survival_comp}). Overall, the Kaplan-Meier survival of Nalm-6 tumor bearing \gls{nsg} mice shown in the \cref{fig:mouse_dosing_ivis_survival} was up to day 40 as reported elsewhere\cite{Fraietta2018}. However, we also included a Kaplan-Meier figure up to day 46 (\cref{fig:mouse_dosing_ivis_survival_full}) where most of the mice euthanized from day 40 through day 46 from \gls{dms} groups showed no or very small fragment of spleen which was due to \gls{gvhd} responses. Similar \gls{gvhd} responses were reported earlier in \gls{nsg} mice where the mice injected with human \gls{pbmc} exhibited acute \gls{gvhd} between \SIrange{40}{50}{\day} post intravenous injection\cite{Ali2012}. Notably, both survival analyses (up to day 40 in \cref{fig:mouse_dosing_ivis_survival} and up to day 46 in \cref{fig:mouse_dosing_ivis_survival_full}) confirmed that \gls{dms}-expanded groups outperformed bead-expanded groups in terms of prolonging survival of Nalm-6 tumor challenged \gls{nsg} mice. Together, these data suggested that \glspl{dms} produce T cells that are not only more potent that bead-expanded T cells (even when accounting for differences in \gls{car} expression) but also showed that \gls{dms} expanded T cells are effective at lower doses. Given the quality control data of the T cells prior to injecting into the mice, it seems that this advantage is either due to the higher \pthp{} or the overall fitness of the T cells given the higher expansion in the case of \gls{dms} (\cref{fig:mouse_dosing_qc_cd4,fig:mouse_dosing_qc_growth}). It was likely not due to the memory phenotype given that it was actually slightly higher in the case of beads (\cref{fig:mouse_dosing_qc_mem}). \subsection{DMS-expanded T cells show greater anti-tumor activity \invivo{} compared to beads} \begin{figure*}[ht!] \begingroup \includegraphics{../figures/mouse_dosing_overview.png} \endgroup \caption[Mouse Dosing Experimental Overview] {Overview of \invivo{} experiment to test \gls{car} T cells expanded with either \glspl{dms} at different doses. } \label{fig:mouse_dosing_overview} \end{figure*} \begin{table}[!h] \centering \caption{Results for \gls{car} T cell \invivo{} dose study} \label{tab:mouse_dosing_results} \input{../tables/mouse_dose_car.tex} \end{table} % TODO put growth first in this figure \begin{figure*}[ht!] \begingroup \includegraphics{../figures/mouse_dosing_qc.png} \phantomsubcaption\label{fig:mouse_dosing_qc_mem} \phantomsubcaption\label{fig:mouse_dosing_qc_cd4} \phantomsubcaption\label{fig:mouse_dosing_qc_growth} \endgroup \caption[Mouse Dosing T cell Characteristics] {Characteristics of T cells harvested at day 14 injected into NSG mice at varying doses. Fractions of T cell subtypes in the day 14 product including \subcap{fig:mouse_dosing_qc_mem}{\ptmemp{}}. \subcap{fig:mouse_dosing_qc_cd4}{\pthp{}}, and \subcap{fig:mouse_dosing_qc_growth}{Fold change of T cells.} } \label{fig:mouse_dosing_qc} \end{figure*} % TODO explain what statistical test was used here \begin{figure*}[ht!] \begingroup \includegraphics{../figures/mouse_dosing_ivis.png} \phantomsubcaption\label{fig:mouse_dosing_ivis_images} \phantomsubcaption\label{fig:mouse_dosing_ivis_plots} \phantomsubcaption\label{fig:mouse_dosing_ivis_survival} \phantomsubcaption\label{fig:mouse_dosing_ivis_survival_comp} \phantomsubcaption\label{fig:mouse_dosing_ivis_survival_full} \endgroup \caption[Mouse Dosing IVIS and Survival Results] {T cells expanded with \glspl{dms} confer greater anti-tumor potency \invivo{} even at lower doses. \subcap{fig:mouse_dosing_ivis_images}{IVIS images of Nalm-6 tumor-bearing \gls{nsg} mice injected with varying doses of T cells} \subcap{fig:mouse_dosing_ivis_plots}{Plots showing quantified photon counts of the results from (\subref{fig:mouse_dosing_ivis_plots}).} \subcap{fig:mouse_dosing_ivis_survival}{Survival plots of mice} \subcap{fig:mouse_dosing_ivis_survival_comp}{Survival plots of mice showing only those that received a comparable number of \gls{car} T cells.} \subcap{fig:mouse_dosing_ivis_survival_full}{The same data as \subref{fig:mouse_dosing_ivis_survival} except showing the full time until euthanasia for all mice (including those that died via \gls{gvhd}).} } \label{fig:mouse_dosing_ivis} \end{figure*} \subsection{Beads and DMSs perform similarly at earlier timepoints} We then asked how T cells harvested using either beads or \gls{dms} performed when harvested at earlier timepoints {\#}{levine paper with early timepoints}. We performed the same experiments as described in \cref{fig:mouse_dosing_overview} with the modification that T cells were only grown and harvested after \SI{6}{\day}, \SI{10}{\day}, or \SI{14}{\day} of expansion (\cref{fig:mouse_timecourse_overview}). T cells were frozen after harvest, and all timepoints were thawed at the same time prior to injection. The dose of T cells injected was \num{1.25e6} cells per mouse (the same as the high dose in the first experiment). All other characteristics of the experiment were the same. \begin{figure*}[ht!] \begingroup \includegraphics{../figures/mouse_timecourse_overview.png} \endgroup \caption[Mouse Timecourse Experimental Overview] {Overview of \invivo{} experiment to test \gls{car} T cells using either \glspl{dms} or bead harvested at varying timepoints. } \label{fig:mouse_timecourse_overview} \end{figure*} % TODO find literature saying that CAR T cells grow slower As was the case with the first \invivo{} experiment, T cells activated with \glspl{dms} expanded much more efficiently compared to those expanded with beads (\cref{fig:mouse_timecourse_qc_growth}). When we quantified the \ptcarp{} of T cells harvested at each timepoint, we noted that the bead group had much higher \ptcar{} expression at earlier timpoints compared to \gls{dms}, while they equalized at later timepoints (\cref{fig:mouse_timecourse_qc_car}). In addition, overall \ptcar{} expression decreased at later timepoints, indicating that \gls{car} transduced T cells either grow slower or die faster compared to untransduced cells. The \pthp{} of the harvested T cells was higher overall in \gls{dms} expanded T cells but decreased with increasing timepoints (\cref{fig:mouse_timecourse_qc_cd4}). The \ptmemp{} was similar at day 6 between bead and \gls{dms} groups but the \gls{dms} group had higher \ptmemp{} at day 14 despite the overall \ptmemp{} decreasing with time as shown elsewhere (\cref{fig:mouse_timecourse_qc_mem}) {\#}{levine paper mem over time}. \begin{figure*}[ht!] \begingroup \includegraphics{../figures/mouse_timecourse_qc.png} \phantomsubcaption\label{fig:mouse_timecourse_qc_growth} \phantomsubcaption\label{fig:mouse_timecourse_qc_car} \phantomsubcaption\label{fig:mouse_timecourse_qc_cd4} \phantomsubcaption\label{fig:mouse_timecourse_qc_mem} \endgroup \caption[Mouse Timecourse T cell Characteristics] {Characteristics of T cells harvested at varying timepoints injected into NSG mice. \subcap{fig:mouse_timecourse_qc_growth}{Fold change of T cells (each timepoint only includes the runs that were harvested at day 14).} Fractions of T cell subtypes in the day 14 product including \subcap{fig:mouse_timecourse_qc_car}{\ptcarp{}}, \subcap{fig:mouse_timecourse_qc_cd4}{\pthp{}}, and \subcap{fig:mouse_timecourse_qc_mem}{\ptmemp{}}. } \label{fig:mouse_timecourse_qc} \end{figure*} We analyzed the tumor burden using \gls{ivis} which showed that mice that received T cells from any group performed better than those that received only saline (\cref{fig:mouse_timecourse_ivis}). Note that unlike the previous experiment, many of the mice survived until day 40 at which point \gls{gvhd} began to take effect (after euthanizing the mice at day 42, most had small or no spleen). When comparing bead and \gls{dms} groups, the \gls{dms} T cells still seemed superior to the bead group, at least initially (note that in this case they had similar numbers of \ptcar{} cells). At day 6, both \gls{dms} and bead groups seemed to eradicate the tumor initially, after which it came back after day 21 for the bead and day 28 for the \gls{dms} group. The day 10 groups performed somewhere in between, where they increased linearly unlike the day 6 groups but not as quickly as the day 14 groups. In the case of the \gls{dms} day 10 group, it also appeared like a few mice actually performed better than all other groups in regard to the final tumor burden. \begin{figure*}[ht!] \begingroup \includegraphics{../figures/mouse_timecourse_ivis.png} \phantomsubcaption\label{fig:mouse_timecourse_ivis_images} \phantomsubcaption\label{fig:mouse_timecourse_ivis_plots} \endgroup \caption[Mouse Timecourse IVIS Results] {\glspl{dms} exhibit superior anti-tumor activity \invivo{} at day 14 compared to beads but are similar to beads at lower timepoints. \subcap{fig:mouse_timecourse_ivis_images}{IVIS images for day 6 to day 42 of mice treated with varying doses of \gls{car} T cells grown with beads or \glspl{dms}.} \subcap{fig:mouse_timecourse_ivis_plots}{Quantified dotplots of the images in (\subref{fig:mouse_timecourse_ivis_images}). Numbers beneath each dot represent the number of mice at that timepoint.}, } \label{fig:mouse_timecourse_ivis} \end{figure*} \section{discussion} % TABLE make a summary table showing the results from both experiments; this is % tough to explain. When we tested bead and DMS expanded \gls{car} T cells, we also found that the \gls{dms} expanded CAR-T cells outperformed bead groups in prolonging survival of Nalm-6 tumor challenged (intravenously injected) \gls{nsg} mice. DMS expanded CAR-T cells were very effective in clearing tumor cells as early as 7 days post CAR-T injection even at low total T cell dose compared to the bead groups where tumor burden was higher than DMS groups across all the total T cell doses tested here. More interestingly, when only CAR-expressing T cell doses between bead and DMS groups were compared, DMS group had significantly higher survival effects over similar or higher CAR expression T cell doses from bead group. All these results suggest that the higher proportion of memory T cells in DMS groups (compared to bead group) resulted in highly effective CAR-T cells that can efficiently kill tumor cells as recently reported in literature\cite{Fraietta2018, Sommermeyer2015}. % TODO try and find literature explaining what the ideal ratio is When testing \gls{car} T cells at earlier timepoints relative to day 14 as used in the first \invivo{} experiment, we noted that none of the \gls{car} treatments seemed to work as well as they did in the first experiment. However, at day 14, we should note that the number of \gls{car} T cells injected in the second experiment was lower than the lowest dose in the first for both bead and \gls{dms} (\cref{fig:mouse_timecourse_qc_car,tab:mouse_dosing_results}). While the \ptmemp{} generally increases with earlier timepoints in the second experiment, the first experiment suggests that \ptmemp{} may not be the primary driver in this particular model (\cref{fig:mouse_timecourse_qc_mem,fig:mouse_dosing_qc_mem}). As with the first experiment, the \pthp{} seems to be higher overall in the \gls{dms} group than the bead group (\cref{fig:mouse_dosing_qc_cd4,fig:mouse_timecourse_qc_cd4}), and this may explain the modest advantage that the \gls{dms} T cells seemed to have in the second experiment in slowing the progression of tumor burden. \chapter{conclusions and future work}\label{conclusions} \section{conclusions} This dissertation describes the development of a novel T cell expansion platform, including the fabrication, quality control, and biological validation of its performance both \invitro{} and \invivo{}. Development of such a system would be meaningful even if it only performed as well as current methods, as adding another method to the arsenal of the growing T cell manufacturing industry would reduce the reliance on a small number of companies that currently license magnetic bead-based T cell expansion technology. However, we additionally show that the \gls{dms} platform expands more T cells on average, including highly potent \ptmem{} and \pth{} T cells, and produces higher percentages of both. If commercialized, this would be a compelling asset the T cell manufacturing industry. % TODO double check the numbers at the end In \cref{aim1}, we develop the \gls{dms} platform and verified its efficacy \invitro{}. Importantly, this included \gls{qc} steps at every critical step of the fabrication process to ensure that the \gls{dms} can be made within a targeted specification. These \gls{qc} steps all rely on common, relatively cost-effective assays such as the \gls{haba} assay, \gls{bca} assay, and \glspl{elisa}, thus other labs and commercial entities should be able to perform them. The microcarriers themselves are an off-the-shelf product available from reputable vendors, further enhancing translatability. On average, we demonstrated that the \gls{dms} outperforms state-of-the-art bead-based T cell expansion technology in terms of total fold expansion, \ptmemp{}, and \pthp{} by \SI{143}{\percent}, \SI{2.5}{\percent}, and \SI{9.8}{\percent} controlling for donor, operator, and a variety of process conditions. In \cref{aim2a}, we developed a modeling pipeline that can be used by commercial entities as the scale up this process to identify \glspl{cqa} and \gls{cpp}. These are highly important for a variety of reasons. First, understanding pertinent \glspl{cpp} allow manufacturers to operate their process at optimal conditions. This is important for anti-tumor cell therapies, where the prospects of a patient can urgently depend on receiving therapy in a timely manner. Optimal process conditions allow T cells to be expanded as quickly as possible for the patient, while also minimizing cost for the manufacturer. Second, \glspl{cqa} can be used to define process control schemes as well as release criteria. Process control, and with it the ability to predict future outcomes based on data obtained at the present, is highly important for cell therapies given that batch failures are extremely expensive {\#}, and predicting a batch failure would allow manufacturers to restart the batch in a timely manner without wasting resources. Furthermore, \glspl{cqa} can be used to define what a `good' vs `bad' product is, which will important help anticipate dosing and followup procedures in the clinic if the T cells are administered. In the aim, we cannot claim to have found the ultimate set of \glspl{cqa} and \glspl{cpp}, as we used tissue culture plates instead of a bioreactor and we only used one donor. However, we have indeed outlined a process that others may use to find these for their process. In particular, the 2-phase modeling process we used (starting with a \gls{doe} and collecting data longitudinally) is a strategy that manufacturers can easily implement. Also, collecting secretome and metabolome is easily generalized to any setting and to most bioreactors and expansion systems, as they can be obtained with relatively inexpensive equipment (Luminex assay, benchtop \gls{nmr}, etc) without disturbing the cell culture. In \cref{aim2b}, we further explored additional tuning knobs that could be used to control and optimize the \gls{dms} system. We determined that altering the \gls{dms} concentration temporally has profound effects on the phenotype and expansion rate. This agrees with other data we obtained in \cref{aim2a} and with what others have generally reported about signal strength and T cell differentiation {\#}. We did not find any mechanistic relationship between either integrin signaling or \gls{il15} signaling. In the case of the former, it may be more likely that the \glspl{dms} surfaces are saturated to the point of sterically hindering any integrin interactions with the collagen surface. In the case of \gls{il15} more experiments likely need to be done in order to plausibly rule out this mechanism and/or determine if it is involved at all. % TODO make this tighter and cite paper showing that this makes at least some % sense In \cref{aim3} we determined that the \glspl{dms} expand T cells that also performed better than beads \invivo{}. In the first experiment we performed, the results were very clearly in favor of the \glspl{dms}. In the second experiment, even the \gls{dms} group failed to fully control the tumor burden, but this is not surprising given the low \ptcarp{} across all groups. Also, despite this, the \gls{dms} group appeared to control the tumor better on average for early, mid, and late T cell harvesting timepoints. It was not clear if this effect was due to increased \cdp{} or overall increased fitness of the \gls{dms}-expanded T cells given their higher expansion rate. The \ptmemp{} did not seem to be a factor given that it was nearly the same in the first experiment between \gls{dms} and bead groups despite the clear advantage seen in the \gls{dms} group. \section{future directions} There are several important next steps to perform with this work: \subsection{Translation to GMP process} While this work was done with translatability and \gls{qc} in mind, an important feature that is missing from the process currently is the use of \gls{gmp} methods and materials. The microcarriers themselves are made from porcine-derived collagen, which itself is not \gls{gmp}-compliant due to its non-human animal origins. However, using any other source of collagen should work so long as the structure of the microcarriers remains relatively similar and it has lysine groups that can react with the \gls{snb} to attach \gls{stp} and \glspl{mab}. Obviously these would need to be tested and verified, but they should not be insurmountable. Furthermore, the \gls{mab} binding step requires \gls{bsa} to prevent adsorption to the non-polar polymer walls of the reaction tubes. A human carrier protein such as \gls{hsa} could be used in its place to eliminate the non-human animal origin material, but this could be much more expensive. Alternatively, the use of protein could be replaced altogether by a non-ionic detergent such as Tween-20 or Tween-80, which are already used for commercial \gls{mab} formulations for precisely this purpose {\#}. Validating the process with Tween would be the best next step to eliminate \gls{bsa} from the process. The \gls{stp} and \glspl{mab} in this process were not \gls{gmp}-grade; however, they are commonly used in clinical technology such as dynabeads and thus the research-grade proteins used here could be easily replaced. The \gls{snb} is a synthetic small molecule and thus does not have any animal-origin concerns. \subsection{Mechanistic investigation} % why do the dms work? % can we put anything on the dms to enhance their potency? \subsection{Assessing performance using unhealthy donors} All the work presented in this dissertation was performed using healthy donors. This was mostly due to the fact that it was much easier to obtain healthy donor cells and was much easier to control. However, it is indisputable that the most relevant test cases of the \gls{dms} will be for unhealthy patient T cells, at least in the case of autologous therapies. In particular, it will be interesting to see how the \gls{dms} performs when assessed head-to-head with bead-based expansion technology given that even in healthy donors, we observed the \gls{dms} platform to work where the beads failed (\cref{fig:dms_exp_fold_change}). \subsection{translation to bioreactors} % use dms in non-static bioreactors such as wave by first activating in a static % environment \onecolumn \clearpage % TODO some people put appendices here....not sure if I need to \chapter{References} \renewcommand{\section}[2]{} % noop the original bib section header \bibliography{references} \bibliographystyle{naturemag} \end{document}