ENH proofread aim2a

This commit is contained in:
Nathan Dwarshuis 2021-08-04 18:24:06 -04:00
parent fa716f9d78
commit 4789723e72
9 changed files with 372 additions and 268 deletions

View File

@ -16,8 +16,8 @@
Observations & 30 \\
R$^{2}$ & 0.888 \\
Adjusted R$^{2}$ & 0.870 \\
Residual Std. Error & 727,042.800 (df = 25) \\
F Statistic & 49.454$^{***}$ (df = 4; 25) \\
% Residual Std. Error & 727,042.800 (df = 25) \\
% F Statistic & 49.454$^{***}$ (df = 4; 25) \\
\hline
\hline \\[-1.8ex]
\textit{Note:} & \multicolumn{1}{r}{$^{*}$p$<$0.1; $^{**}$p$<$0.05; $^{***}$p$<$0.01} \\

View File

@ -16,8 +16,8 @@
Observations & 30 \\
R$^{2}$ & 0.331 \\
Adjusted R$^{2}$ & 0.224 \\
Residual Std. Error & 3,659,501.000 (df = 25) \\
F Statistic & 3.096$^{**}$ (df = 4; 25) \\
% Residual Std. Error & 3,659,501.000 (df = 25) \\
% F Statistic & 3.096$^{**}$ (df = 4; 25) \\
\hline
\hline \\[-1.8ex]
\textit{Note:} & \multicolumn{1}{r}{$^{*}$p$<$0.1; $^{**}$p$<$0.05; $^{***}$p$<$0.01} \\

View File

@ -23,8 +23,8 @@
Observations & 30 \\
R$^{2}$ & 0.741 \\
Adjusted R$^{2}$ & 0.583 \\
Residual Std. Error & 0.228 (df = 18) \\
F Statistic & 4.693$^{***}$ (df = 11; 18) \\
% Residual Std. Error & 0.228 (df = 18) \\
% F Statistic & 4.693$^{***}$ (df = 11; 18) \\
\hline
\hline \\[-1.8ex]
\textit{Note:} & \multicolumn{1}{r}{$^{*}$p$<$0.1; $^{**}$p$<$0.05; $^{***}$p$<$0.01} \\

View File

@ -14,8 +14,8 @@
Observations & 30 \\
R$^{2}$ & 0.835 \\
Adjusted R$^{2}$ & 0.808 \\
Residual Std. Error & 493,168.700 (df = 25) \\
F Statistic & 31.571$^{***}$ (df = 4; 25) \\
% Residual Std. Error & 493,168.700 (df = 25) \\
% F Statistic & 31.571$^{***}$ (df = 4; 25) \\
\hline
\hline \\[-1.8ex]
\textit{Note:} & \multicolumn{1}{r}{$^{*}$p$<$0.1; $^{**}$p$<$0.05; $^{***}$p$<$0.01} \\

View File

@ -15,8 +15,8 @@
Observations & 30 \\
R$^{2}$ & 0.879 \\
Adjusted R$^{2}$ & 0.860 \\
Residual Std. Error & 0.039 (df = 25) \\
F Statistic & 45.554$^{***}$ (df = 4; 25) \\
% Residual Std. Error & 0.039 (df = 25) \\
% F Statistic & 45.554$^{***}$ (df = 4; 25) \\
\hline
\hline \\[-1.8ex]
\textit{Note:} & \multicolumn{1}{r}{$^{*}$p$<$0.1; $^{**}$p$<$0.05; $^{***}$p$<$0.01} \\

View File

@ -1,7 +1,7 @@
\begin{tabular}{ccccc}
\\[-1.8ex]\hline
\hline \\[-1.8ex]
\\[-1.8ex] Dataset & Run & IL2 Conc (\si{\IU\per\ml}) & DMS Conc. (\si{\IU\per\ml})
\\[-1.8ex] Dataset & Run & IL2 Conc (\si{\IU\per\ml}) & DMS Conc. (\si{\dms\per\ml})
& Functional \gls{mab} (\si{\percent}) \\
\hline \\[-1.8ex]
DOE & 1 & 30 & 500 & 100\\

View File

@ -17,7 +17,7 @@ PP+S6 & \SI{98}{\percent} & \SI{71.4}{\percent} & \SI{99.9}{\percent} & \SI{75.0
PP+S6+N6 & \SI{98}{\percent} & \SI{68.2}{\percent} & \SI{95.6}{\percent} & \SI{74.4}{\percent} & \SI{72.5}{\percent} & \SI{81.7}{\percent} & \SI{77.0}{\percent}\\
\hline \\
\multicolumn{8}{l}{\ptmemh{} cells} \\
\multicolumn{8}{l}{\ptmemk{} cells} \\
PP+N4 & \SI{93}{\percent} & \SI{4.7}{\percent} & \SI{44.4}{\percent} & \SI{9.2}{\percent} & \SI{1.2}{\percent} & \SI{65.1}{\percent} & \SI{9.1}{\percent}\\
PP+N6 & \SI{86}{\percent} & \SI{2.0}{\percent} & \SI{29.9}{\percent} & \SI{15.8}{\percent} & \SI{28.5}{\percent} & \SI{63.3}{\percent} & \SI{30.6}{\percent}\\
PP+S6 & \SI{93}{\percent} & \SI{7.8}{\percent} & \SI{28.0}{\percent} & \SI{15.1}{\percent} & \SI{76.2}{\percent} & \SI{98.4}{\percent} & \SI{49.8}{\percent}\\

View File

@ -2673,6 +2673,62 @@ CONCLUSIONS: We developed a simplified, semi-closed system for the initial selec
publisher = {The American Association of Immunologists},
}
@Article{Delaglio1995,
author = {Frank Delaglio and Stephan Grzesiek and GeertenW. Vuister and Guang Zhu and John Pfeifer and Ad Bax},
journal = {Journal of Biomolecular {NMR}},
title = {{NMRPipe}: A multidimensional spectral processing system based on {UNIX} pipes},
year = {1995},
month = {nov},
number = {3},
volume = {6},
doi = {10.1007/bf00197809},
publisher = {Springer Science and Business Media {LLC}},
}
@Article{Dieterle2006,
author = {Frank Dieterle and Alfred Ross and Götz Schlotterbeck and Hans Senn},
journal = {Analytical Chemistry},
title = {Probabilistic Quotient Normalization as Robust Method to Account for Dilution of Complex Biological Mixtures. Application in1H {NMR} Metabonomics},
year = {2006},
month = {jul},
number = {13},
pages = {4281--4290},
volume = {78},
doi = {10.1021/ac051632c},
publisher = {American Chemical Society ({ACS})},
}
@Article{Dashti2017,
author = {Hesam Dashti and William M. Westler and Marco Tonelli and Jonathan R. Wedell and John L. Markley and Hamid R. Eghbalnia},
journal = {Analytical Chemistry},
title = {Spin System Modeling of Nuclear Magnetic Resonance Spectra for Applications in Metabolomics and Small Molecule Screening},
year = {2017},
month = {nov},
number = {22},
pages = {12201--12208},
volume = {89},
doi = {10.1021/acs.analchem.7b02884},
publisher = {American Chemical Society ({ACS})},
}
@InProceedings{Kordona,
author = {A.K. Kordon and Ching-Tai Lue},
booktitle = {Proceedings of the 2004 Congress on Evolutionary Computation ({IEEE} Cat. No.04TH8753)},
title = {Symbolic regression modeling of blown film process effects},
year = {2004},
publisher = {{IEEE}},
doi = {10.1109/cec.2004.1330907},
}
@Book{Koza1992,
author = {Koza, John},
publisher = {MIT Press},
title = {Genetic programming : on the programming of computers by means of natural selection},
year = {1992},
address = {Cambridge, Mass},
isbn = {0262111705},
}
@Comment{jabref-meta: databaseType:bibtex;}
@Comment{jabref-meta: grouping:

View File

@ -103,8 +103,9 @@
\tcellacronym{tc}{c}{cytotoxic}{}
\tcellacronym{th1}{h}{type 1 helper}{1}
\tcellacronym{th2}{h}{type 2 helper}{2}
% \tcellacronym{th17}{h}{\il{17} helper}{1}
\tcellacronym{th17}{h}{IL-17 helper}{17}
\newacronym{bcaa}{BCAA}{branched-chain amino acid}
\newacronym{til}{TIL}{tumor infiltrating lymphocyte}
\newacronym{tcr}{TCR}{T cell receptor}
\newacronym{act}{ACT}{adoptive cell therapies}
@ -207,6 +208,7 @@
\newacronym{scfv}{scFv}{single-chain fragment variable}
\newacronym{hepes}{HEPES}{4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid}
\newacronym{nhs}{NHS}{N-hydroxysulfosuccinimide}
\newacronym{tocsy}{TOCSY}{total correlation spectroscopy}
%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%%
% SI units for uber nerds
@ -282,10 +284,13 @@
% so I don't need to worry about abbreviating all the different interleukins
\newcommand{\il}[1]{\gls{il}-#1}
% DOE responses I don't feel like typing ad-nauseam
% DOE stuff 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}
\newcommand{\rmemh}{total \ptmemh{} cells}
\newcommand{\rmemk}{total \ptmemk{} cells}
\newcommand{\rratio}{CD4/CD8 ratio}
% vendor and product stuff I don't feel like typing
\newcommand{\catnum}[2]{(#1, #2)}
@ -2683,12 +2688,12 @@ necessary to have a fully-automated manufacturing system.
The \gls{dms} system could be used as a drop in replacement for beads in many of
current allogeneic therapies. Indeed, given its higher potential for expansion
(\cref{fig:dms_exp,tab:ci_controlled}, it may work in cases where the beads fail
(although this would need to be tested by gathering data with many unhealthy
donors). However, in the autologous setting patients only need a fixed dose, and
thus any expansion beyond the indicated dose would be wasted. Given this, it
will be interesting to apply this technology in an allogeneic paradigm where
this increased expansion potential would be well utilized.
(\cref{fig:dms_exp,tab:ci_controlled}), it may work in cases where the beads
fail (although this would need to be tested by gathering data with many
unhealthy donors). However, in the autologous setting patients only need a fixed
dose, and thus any expansion beyond the indicated dose would be wasted. Given
this, it will be interesting to apply this technology in an allogeneic paradigm
where this increased expansion potential would be well utilized.
Finally, we should note that while we demonstrated a method providing superior
performance compared to bead-based expansion, the cell manufacturing field would
@ -2702,20 +2707,20 @@ manufacturing companies.
\section{introduction}
The purpose of this sub-aim was to develop computational methods to identify novel
\glspl{cqa} and \glspl{cpp} that could be used for release criteria, process
control, and process optimization for the \gls{dms} platform. We hypothesized
that T cells grown using the \gls{dms} system would produce detectable
biological signatures in the media supernatent which corresponded to clinically
relevent responses such as the fold expansion of the T cells or the resulting
phenotype. We tested this hypothesis by activating T cells under a variety of
conditions using a \gls{doe}, sampling the media at intermediate timepoints, and
creating models to predict the outcome of the cultures. We should stress that
the specific \glspl{cpp} and \glspl{cqa} determined by this aim are not
necessarily universal, as this was not performed with equipment that would
normally be used at scale. However, the process outlined here is one that can
easily be adaptable to any system, and the specific findings themselves offer
interesting insights that warrant further study.
The purpose of this sub-aim was to develop computational methods to identify
novel \glspl{cqa} and \glspl{cpp} that could be used for release criteria,
process control, and process optimization for the \gls{dms} platform. We
hypothesized that T cells grown using the \gls{dms} system would produce
detectable biological signatures in the media supernatent which corresponded to
clinically relevent responses such as fold expansion or phenotype. We tested
this hypothesis by activating T cells under a variety of conditions using a
\gls{doe}, sampling the media at intermediate timepoints, and creating models to
predict the outcome of the cultures. We should stress that the specific
\glspl{cpp} and \glspl{cqa} determined by this aim are not necessarily
universal, as this was not performed with equipment that would normally be used
at scale. However, the process outlined here is one that can easily be adaptable
to any system, and the specific findings themselves offer interesting insights
that warrant further study.
\section{methods}
@ -2744,33 +2749,38 @@ interesting insights that warrant further study.
\label{fig:mod_overview}
\end{figure*}
The first DOE resulted in a randomized 18-run I-optimal custom design where each
DMS parameter was evaluated at three levels: IL2 concentration (10, 20, and 30
U/uL), DMS concentration (500, 1500, 2500 carrier/uL), and functionalized
antibody percent (60\%, 80\%, 100\%). These 18 runs consisted of 14 unique
parameter combinations where 4 of them were replicated twice to assess
prediction error. Process parameters for the ADOE were evaluated at multiple
levels: IL2 concentration (30, 35, and 40 U/uL), DMS concentration (500, 1000,
1500, 2000, 2500, 3000, 3500 carrier/uL), and functionalized antibody percent
(100\%) as depicted in Fig.1b. To further optimize the initial region explored
(DOE) in terms of total live CD4+ TN+TCM cells, a sequential adaptive
design-of-experiment (ADOE) was designed with 10 unique parameter combinations,
two of these replicated twice for a total of 12 additional samples (Fig.1b). The
fusion of cytokine and NMR profiles from media to model these responses included
30 cytokines from a custom Thermo Fisher ProcartaPlex Luminex kit and 20 NMR
features. These 20 spectral features from NMR media analysis were selected out
of approximately 250 peaks through the implementation of a variance-based
feature selection approach and some manual inspection steps.
The overall workflow of this aim is shown in \cref{fig:mod_overview_flow}.
Experimental conditions within the design space were explored using a \gls{doe},
and longitudinal samples were collected for each condition as the cultures
progressed. Data from inputs and/or longitudinal samples were used to predict
the endpoint response. The fusion of cytokine and \gls{nmr} profiles from media
to model these responses included 30 cytokines from a custom Thermo Fisher
ProcartaPlex Luminex kit and 20 \gls{nmr} features. These 20 spectral features
from \gls{nmr} media analysis were selected out of approximately 250 peaks
through the implementation of a variance-based feature selection approach and
some manual inspection steps.
The first \gls{doe} resulted in a randomized 18-run I-optimal custom design
where each \gls{dms} parameter was evaluated at three levels: \pilII{} (10, 20,
and 30 U/uL), \pdms{} (500, 1500, 2500 \si{\dms\per\ul}), and \pmab{} (60, 80,
100 \si{\percent}). These 18 runs consisted of 14 unique parameter combinations
where 4 of them were replicated twice to assess prediction error. To further
optimize the initial region explored, an \gls{adoe} was designed with 10 unique
parameter combinations, two of these replicated twice for a total of 12
additional samples (\cref{fig:mod_overview_doe}). Process parameters for the
\gls{adoe} were evaluated at multiple levels: \pilII{} (30, 35, and 40
\si{\IU\per\ml}), \pdms{} (500, 1000, 1500, 2000, 2500, 3000, 3500
\si{\dms\per\ml}), and \pmab{} (\SI{100}{\percent}) (\cref{fig:mod_overview}).
\subsection{DMS fabrication}
\glspl{dms} were fabricated as described in \cref{sec:dms_fab} with the
following modifications in order to obtain a variable functional \gls{mab}
surface density. During the \gls{mab} coating step, the anti-CD3/anti-CD28 mAb
mixture was further combined with a biotinylated isotype control to reduce the
overall fraction of targeted \glspl{mab} (for example the \SI{60}{\percent}
\gls{mab} surface density corresponded to 3 mass parts \acd{3}, 3 mass parts
\acd{28}, and 4 mass parts isotype control).
surface density. During the \gls{mab} coating step, the \acd{3}/\acd{28}
\gls{mab} mixture was further combined with a biotinylated isotype control to
reduce the overall fraction of targeted \glspl{mab} (for example the
\SI{60}{\percent} \gls{mab} surface density corresponded to 3 mass parts
\acd{3}, 3 mass parts \acd{28}, and 4 mass parts isotype control).
\subsection{T cell culture}
@ -2779,9 +2789,9 @@ following modifications. At days 4, 6, 8, and 11, \SI{100}{\ul} media were
collected for the Luminex assay and \gls{nmr} analysis. The volume of removed
media was equivalently replaced during the media feeding step, which took place
immediately after sample collection. Additionally, the same media feeding
schedule was followed for the DOE and ADOE to improve consistency, and the same
donor lot was used for both experiments. All cell counts were performed using
\gls{aopi}.
schedule was followed for the \gls{doe} and \gls{adoe} to improve consistency,
and the same donor lot was used for both experiments. All cell counts were
performed using \gls{aopi}.
\subsection{flow cytometry}
@ -2796,26 +2806,29 @@ Cytokines were quantified via Luminex as described in
Prior to analysis, samples were centrifuged at \SI{2990}{\gforce} for
\SI{20}{\minute} at \SI{4}{\degreeCelsius} to clear any debris\footnote{all
\gls{nmr} analysis was done by our collaborators at the University of
Georgia}. 5 uL of 100/3 mM DSS-D6 in deuterium oxide (Cambridge Isotope
Laboratories) were added to 1.7 mm NMR tubes (Bruker BioSpin), followed by 45 uL
of media from each sample that was added and mixed, for a final volume of 50 uL
in each tube. Samples were prepared on ice and in predetermined, randomized
order. The remaining volume from each sample in the rack (approx. 4 uL) was
combined to create an internal pool. This material was used for internal
controls within each rack as well as metabolite annotation.
\gls{nmr} analysis was done by our collaborators Max Colonna and Art Edison at
the University of Georgia; methods included here for reference}. \SI{5}{\ul} of
100/3 \si{\mM} DSS-D6 in deuterium oxide (Cambridge Isotope Laboratories) were
added to \SI{1.7}{\mm} \gls{nmr} tubes (Bruker BioSpin), followed by
\SI{45}{\ul} of media from each sample that was added and mixed, for a final
volume of \SI{50}{\ul} in each tube. Samples were prepared on ice and in
predetermined, randomized order. The remaining volume from each sample in the
rack (approx. \SI{4}{\ul}) was combined to create an internal pool. This
material was used for internal controls within each rack as well as metabolite
annotation.
\gls{nmr} spectra were collected on a Bruker Avance III HD spectrometer at 600
MHz using a 5-mm TXI cryogenic probe and TopSpin software (Bruker BioSpin).
One-dimensional spectra were collected on all samples using the noesypr1d pulse
sequence under automation using ICON NMR software. Two-dimensional \gls{hsqc}
and TOCSY spectra were collected on internal pooled control samples for
metabolite annotation.
\gls{nmr} spectra were collected on a Bruker Avance III HD spectrometer at
\SI{600}{\MHz} using a \SI{5}{\mm} TXI cryogenic probe and TopSpin software
(Bruker BioSpin). One-dimensional spectra were collected on all samples using
the noesypr1d pulse sequence under automation using ICON NMR software.
Two-dimensional \gls{hsqc} and \gls{tocsy} spectra were collected on internal
pooled control samples for metabolite annotation.
One-dimensional spectra were manually phased and baseline corrected in TopSpin.
Two-dimensional spectra were processed in NMRpipe37. One dimensional spectra
were referenced, water/end regions removed, and normalized with the PQN
algorithm38 using an in-house MATLAB (The MathWorks, Inc.) toolbox.
Two-dimensional spectra were processed in NMRpipe\cite{Delaglio1995}. One
dimensional spectra were referenced, water/end regions removed, and normalized
with the PQN algorithm\cite{Dieterle2006} using an in-house MATLAB (The
MathWorks, Inc.) toolbox.
% TODO add the supplemental figure alluded to here?
To reduce the total number of spectral features from approximately 250 peaks and
@ -2824,19 +2837,22 @@ variance-based feature selection was performed within MATLAB. For each digitized
point on the spectrum, the variance was calculated across all experimental
samples and plotted. Clearly-resolved features corresponding to peaks in the
variance spectrum were manually binned and integrated to obtain quantitative
feature intensities across all samples (Supp.Fig.S24). In addition to highly
variable features, several other clearly resolved and easily identifiable
features were selected (glucose, BCAA region, etc). Some features were later
discovered to belong to the same metabolite but were included in further
analysis.
feature intensities across all samples.
% (Supp.Fig.S24).
In addition to highly variable features, several other clearly resolved and
easily identifiable features were selected (glucose, \gls{bcaa} region, etc).
Some features were later discovered to belong to the same metabolite but were
included in further analysis.
% I think this is the right source? it seems wrong in the manuscript but this
% source at least talks about an optimization score
Two-dimensional spectra collected on pooled samples were uploaded to COLMARm web
server10, where \gls{hsqc} peaks were automatically matched to database peaks.
server, where \gls{hsqc} peaks were automatically matched to database peaks.
\gls{hsqc} matches were manually reviewed with additional 2D and proton spectra
to confirm the match. Annotations were assigned a confidence score based upon
the levels of spectral data supporting the match as previously described11.
Annotated metabolites were matched to previously selected features used for
statistical analysis.
the levels of spectral data supporting the match as previously
described\cite{Dashti2017}. Annotated metabolites were matched to previously
selected features used for statistical analysis.
% I'm pretty sure this isn't relevant
% Using the list of annotated metabolites obtained above, an approximation of a
@ -2851,8 +2867,8 @@ statistical analysis.
Several low abundance features selected for analysis did not have database
matches and were not annotated. Statistical total correlation spectroscopy41
suggested that some of these unknown features belonged to the same molecules
(not shown). Additional multidimensional NMR experiments will be required to
determine their identity.
(not shown). Additional multidimensional \gls{nmr} experiments will be required
to determine their identity.
\subsection{machine learning and statistical analysis}
@ -2861,66 +2877,70 @@ Linear regression analysis of the \glspl{doe} was performed as described in
Seven \gls{ml} techniques were implemented to predict three responses related to
the memory phenotype of the cultured T cells under different process parameters
conditions (i.e. Total Live CD4+ TN and TCM, Total Live CD8+ TN+TCM, and Ratio
CD4+/CD8+ TN+TCM). The \gls{ml} methods executed were \gls{rf}, \gls{gbm},
\gls{cif}, \gls{lasso}, \gls{plsr}, \gls{svm}, and DataModelers
\gls{sr}\footnote{of these seven methods, all except \gls{lasso} were performed
by collaborators}. Primarily, \gls{sr} models were used to optimize process
parameter values based on TN+TCM phenotype and to extract early predictive
conditions (i.e. \rmemh{}, \rmemk{}, and \rratio{}). The \gls{ml} methods
executed were \gls{rf}, \gls{gbm}, \gls{cif}, \gls{lasso}, \gls{plsr},
\gls{svm}, and DataModelers \gls{sr}\footnote{\gls{sr} was performed by Theresa
Kotanchek at Evolved Analytics, \gls{rf}, \gls{gbm}, \gls{cif}, \gls{plsr},
\gls{svm} were performed by Valerie Odeh-Couvertier at UPRM. Methods included
here for reference}. Primarily, \gls{sr} models were used to optimize process
parameter values based on \ptmem{} phenotype and to extract early predictive
variable combinations from the multi-omics experiments. Furthermore, all
regression methods were executed, and the high-performing models were used to
perform a consensus analysis of the important variables to extract potential
critical quality attributes and critical process parameters predictive of T-cell
critical quality attributes and critical process parameters predictive of T cell
potency, safety, and consistency at the early stages of the manufacturing
process.
\gls{sr} was done using Evolved Analytics DataModeler software (Evolved
Analytics LLC, Midland, MI). DataModeler utilizes genetic programming to evolve
symbolic regression models (both linear and non-linear) rewarding simplicity and
accuracy. Using the selection criteria of highest accuracy (R2>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, DataModelers 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
accuracy. Using the selection criteria of highest accuracy
($R^2$>\SI{90}{\percent}) 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, DataModelers
\inlinecode{SymbolicRegression} function was used to develop explicit algebraic
(linear and nonlinear) models. The fittest models were analyzed to identify the
dominant variables using the \inlinecode{VariablePresence} function, the
dominant variable combinations using the \inlinecode{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, \gls{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.
\inlinecode{ModelDimensionality} function. \inlinecode{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, \gls{anova} tables,
model prediction plot, exportable model forms) using the
\inlinecode{ModelSummaryTable} function. Ensemble prediction and residual
performance were assessed via the \inlinecode{EnsemblePredictionPlot} and
\inlinecode{EnsembleResidualPlot} subroutines respectively. Model maxima
(\inlinecode{ModelMaximum} function) and model minima (\inlinecode{ModelMinimum}
function) were calculated and displayed using the
\inlinecode{ResponsePlotExplorer} function. Trade-off between multiple
responses was explored using \inlinecode{MultiTargetResponseExplorer} and
\inlinecode{ResponseComparisonExplorer} with additional insights derived from
\inlinecode{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
\inlinecode{randomForest}, inlinecode{gbm}, and \inlinecode{cforest} regression
functions in R, for \gls{rf}, \gls{gbm}, and \gls{cif} models, respectively.
Both \gls{rf} and \gls{cif} 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
training stage. \gls{rf} 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, \gls{gbm} 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 \gls{mse}). Prediction performance was
evaluated using \gls{loocv} and permutation-based
variable importance scores assessing \% increase of \gls{mse}, relative
influence based on the increase of prediction error, coefficient values for
\gls{rf}, \gls{gbm}, and \gls{cif}, respectively. \gls{plsr} was executed using
the \inlinecode{plsr} function from the \inlinecode{pls} package in R while
\gls{lasso} regression was performed using the \inlinecode{cv.glmnet} R package,
both using leave-one-out cross-validation. Finally, the \inlinecode{kernlab} R
package was used to construct the \gls{svm} models.
of previous regression trees in an iterative fashion to correct errors
(\gls{mse}) from its precursors trees. Prediction performance was evaluated
using \gls{loocv} and permutation-based variable importance scores assessing
percent increase of \gls{mse}, relative influence based on the increase of
prediction error, coefficient values for \gls{rf}, \gls{gbm}, and \gls{cif},
respectively. \gls{plsr} was executed using the \inlinecode{plsr} function from
the \inlinecode{pls} package in R while \gls{lasso} regression was performed
using the \inlinecode{cv.glmnet} R package, both using \gls{loocv}. Finally, the
\inlinecode{kernlab} R package was used to construct the \gls{svm} models.
Parameter tuning was done for all models in a grid search manner using the train
function from the \inlinecode{caret} R package using \gls{loocv} as the
@ -2936,12 +2956,12 @@ optimum \gls{loocv} performance as well. For \gls{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 \inlinecode{selectNcomp} from
the \inlinecode{pls} package. Moreover, \gls{lasso} regression was performed
using the \inlinecode{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. \inlinecode{svmLinear}) was used to build the \gls{svm}
regression models evaluating the cost parameter value with best \gls{loocv}.
Prediction performance was measured for all models using the final model with
\gls{loocv} tuned parameters.
using the \inlinecode{cv.glmnet} package with $\upalpha$ = 1. The best
$\uplambda$ for each response was chosen using the minimum error criteria.
Lastly, a fixed linear kernel (\inlinecode{svmLinear}) was used to build
the \gls{svm} regression models evaluating the cost parameter value with best
\gls{loocv}. Prediction performance was measured for all models using the final
model with \gls{loocv} tuned parameters.
% TODO do I need this?
% Table M2 shows the parameter values evaluated per model
@ -2961,16 +2981,17 @@ values, absolute coefficient values, weighted sum of absolute coefficients
values, and relative importance from sensitivity analysis determined for
\gls{rf}, \gls{gbm}, \gls{cif}, \gls{lasso}, \gls{plsr}, and \gls{svm},
respectively. Using these scores, key predictive variables were selected if
their importance scores were within the 80th percentile ranking for the
their importance scores were within the \nth{80} percentile ranking for the
following \gls{ml} methods: \gls{rf}, \gls{gbm}, \gls{cif}, \gls{lasso},
\gls{plsr}, \gls{svm} while for \gls{sr} variables present in >30\% of the
top-performing \gls{sr} models from DataModeler (R2>= 90\%, Complexity >= 100)
were chosen to investigate consensus except for \gls{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 \gls{ml}
models) and depicted using a Venn diagram from the \inlinecode{venn} R package.
\gls{plsr}, \gls{svm} while for \gls{sr} variables present in >\SI{30}{\percent}
of the top-performing \gls{sr} models from DataModeler ($R^2\ge$
\SI{90}{\percent}, Complexity $\ge$ 100) were chosen to investigate consensus
except for \gls{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 >\SI{40}{\percent} of the best performing models. Only
variables with those high percentile scoring values were evaluated in terms of
their logical relation (intersection across \gls{ml} models) and depicted using
a Venn diagram from the \inlinecode{venn} R package.
\section{results}
@ -3033,8 +3054,6 @@ 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.
% RESULT this is not consistent with the next section since the responses are
% different
\subsection{DOE shows optimal conditions for expanded potent T cells}
% TABLE not all of these were actually used, explain why by either adding columns
@ -3045,7 +3064,6 @@ at \SI{10}{\IU\per\ml} throughout the remainder of this aim.
\input{../tables/doe_runs.tex}
\end{table}
% RESULT integrate this figure into the results paragraph
\begin{figure*}[ht!]
\begingroup
@ -3183,7 +3201,7 @@ 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
this response; the $R^2$ was 0.741 but the $R_{adj}^2$ 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.
@ -3193,7 +3211,7 @@ 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
All these models appeared to fit will, with $R^2$ and $R_{adj}^2$ 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
@ -3216,16 +3234,24 @@ significant predictors.
\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.
We then visualized the total \ptmemh{} cells and \rratio{} 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_sr_contour}). 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}
\subsection{Modeling with artificial intelligence methods reveals potential
CQAs}
Due to the heterogeneity of the multivariate data collected and knowing that no
single model is perfect for all applications, we implemented an agnostic
modeling approach to better understand these \ptmem{} responses. To achieve
this, a consensus analysis using seven \gls{ml} techniques, \gls{rf}, \gls{gbm},
\gls{cif}, \gls{lasso}, \gls{plsr}, \gls{svm}, and DataModelers \gls{sr}, was
implemented to molecularly characterize \ptmem{} cells and to extract predictive
features of quality early in their expansion process.
\begin{figure*}[ht!]
\begingroup
@ -3238,34 +3264,38 @@ in the model.
\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 DataModelers
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).
We collected secretome data via luminex for days 4, 6, 8, 11, and 14.
Plotting the concentrations of these cytokines showed a large variation over all
runs and between different timepoints, demonstrated that these could potentially
be used to differentiate between different process conditions qualitatively
simply based on variance (\cref{fig:doe_luminex}). These were also much higher
in most cases that a set of bead based runs which were run in parallel, in
agreement with the luminex data obtained previously in the Grex system (these
data were collected in plates) (\cref{fig:grex_luminex}).
% TABLE this table looks like crap, break it up into smaller tables
\begin{table}[!h] \centering
\caption{Results for data-driven modeling}
\caption{Results for data-driven modeling using process parameters (PP) with
only \gls{nmr} on day 4 (N4), only \gls{nmr} on day 6 (N6), only secretome
on day 6 (S6), or various combindation of each for all seven \gls{ml}
techniques}
\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.
\gls{sr} models achieved the highest predictive performance
($R^2$>\SI{93}{\percent}) when using multi-omics predictors for all endpoint
responses (\cref{tab:mod_results}). \gls{sr} achieved $R^2$>\SI{98}{\percent}
while \gls{gbm} tree-based ensembles showed \gls{loocv} $R^2$ >
\SI{95}{\percent} for \rmemh{} and \rmemk{} responses. Similarly, \gls{lasso},
\gls{plsr}, and \gls{svm} methods showed consistently high \gls{loocv},
(\SI{92.9}{\percent}, \SI{99.7}{\percent}, and \SI{90.5}{\percent}
respectively), to predict the \rratio{}. Yet, about \SI{10}{\percent} reduction
in \gls{loocv}, \SIrange{72.5}{81.7}{\percent}, was observed for \rmemh{} with
these three methods. Lastly, \gls{sr} and \gls{plsr} achieved
$R^2$>\SI{90}{\percent} while other \gls{ml} methods exhibited exceedingly
variable \gls{loocv} (\SI{0.3}{\percent} for \gls{rf} to \SI{51.5}{\percent} for
\gls{lasso}) for \rmemk{}.
\begin{figure*}[ht!]
\begingroup
@ -3279,13 +3309,12 @@ cells.
\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}).
The top-performing technique, \gls{sr}, showed that the median aggregated
predictions for \rmemh{} \rmemk{} increases when IL2 concentration, IL15, and
IL2R increase while IL17a decreases in conjunction with other features. These
patterns combined with low values of \pdms{} and GM-CSF uniquely characterized
maximum \rmemk{}. Meanwhile, higher glycine but lower IL13 in combination with
others showed maximum \rmemh{} predictions (\cref{fig:sr_omics}).
\begin{figure*}[ht!]
\begingroup
@ -3306,13 +3335,25 @@ combination with others showed maximum CD4+ TN+TCM predictions
\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).
Selecting \gls{cpp} and \glspl{cqa} candidates consistently for T cell memory is
desired. Here, \gls{tnfa} was found in consensus across all seven \gls{ml}
methods for predicting \rratio{} when considering features with the highest
importance scores across models (\cref{fig:mod_flower_48r}). Other features,
IL2R, IL4, IL17a, and \pdms{}, were commonly selected in $\ge$ 5 \gls{ml}
methods (\cref{fig:mod_flower_48r}). When restricting the models only to include
metabolome, formate emerged as the dominant predictor shared across all seven
models.
% Moreover, IL13 and IL15 were found predictive in combination
% with these using \gls{sr} (Supp.Table.S4).
When performing similar analysis on \rmemh{}, we observe that no species for
either the secretome or metabolome was agreed upon by all seven models
(\cref{fig:mod_flower_cd4}). We also observed that these models did not fit as
well as they did for \rratio{} (\cref{tab:mod_results}). For the secretome, the
species that were agreed upon by $\ge$ 5 models were IL4, IL17a, and IL2R. For
the metabolome, formate once again was agreed upon by $\ge$ 5 models as well as
lactate.
\begin{figure*}[ht!]
\begingroup
@ -3338,55 +3379,61 @@ IL13 and IL15 were found predictive in combination with these using SR
\label{fig:nmr_cors}
\end{figure*}
We also investigated the \gls{nmr} features extracted from day of expansion to
assess if there was any predictive power for \ptmemh{}; in general these models
had almost as good of fit despite being 2 days earlier in the process
(\cref{fig:nmr_cors}). Lactate and formate were observed to correlate with each
other, and both correlated with \rmemh{}. Furthermore, lactate was observed to
positively correlate with \pdms{} and negatively correlate with glucose
(\cref{fig:nmr_cors_lactate}). Formate also had the same correlation patterns
(\cref{fig:nmr_cors_formate}). Glucose was only negatively correlated with
formate and lactate (\cref{fig:nmr_cors_glucose}). Together, these data suggest
that lactate, formate, \pdms{}, and \rmemh{} are fundamentally linked.
\section{discussion}
% optimization of process features
% TODO this sounds like total fluff
% DISCUSSION integrate figures
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
\gls{cpp} 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 \gls{doe} datasets. \gls{sr} has the necessary
capabilities to resolve the issues of process effects modeling and has been
applied across multiple industries\cite{Kordona}. \gls{sr} discovers
mathematical expressions that fit a given sample and differs from conventional
regression techniques in that a model structure is not defined a
priori\cite{Koza1992}. Hence, a key advantage of this methodology is that
transparent, human-interpretable models can be generated from small and large
datasets with no prior assumptions\cite{Kotancheka}.
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 located\cite{Kotancheka}.
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 understanding\cite{Kotancheka}.
competitive 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 located\cite{Kotancheka}. Exploiting these features
allows adaptive data collection and interactive modeling. Consequently, this
\gls{adoe} 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 understanding\cite{Kotancheka}.
% 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).
An in-depth characterization of potential \gls{dms} based T cell \glspl{cqa}
includes a list of cytokine and \gls{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 \gls{nmr} media analysis and process parameters
(\cref{fig:mod_flower}).
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.
\rratio{} 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 cells\cite{Witkowska2005}. Since IL2R was much greater
@ -3399,22 +3446,23 @@ form, this may either increase or decrease CD4+ ratio and/or memory T cells
depending on the ratio of the membrane to soluble TNF\cite{Mehta2018}. 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 population\cite{Wong2011}. This cytokine
has limited signaling in T cells and is thought to be more of an effector than a
differentiation cytokine\cite{Junttila2018}. 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 cells\cite{Amatya2017} 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
for \gls{th2} response and therefore could be secreted if there are significant
\glspl{th2} already present in the starting population\cite{Wong2011}. This
cytokine has limited signaling in T cells and is thought to be more of an
effector than a differentiation cytokine\cite{Junttila2018}. It might be
emerging as relevant due to an initially large number of \glspl{th2} or because
\glspl{th2} were preferentially expanded; indeed, IL4, also found important, is
the conical cytokine that induces \gls{th2} differentiation
(\cref{fig:mod_flower}). The role of these cytokines could be investigated by
quantifying \glspl{th1}, \glspl{th2}, or \glspl{th17} both in the starting
population and longitudinally. Similar to IL13, IL17 is an effector cytokine
produced by \glspl{th17}\cite{Amatya2017} thus may reflect the number of
\glspl{th17} in the population. GM-CSF has been linked with activated T cells,
specifically \glspl{th17}, 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 expansion\cite{Becher2016}. 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
been shown to be essential for memory signaling and effective in skewing
\gls{car} T cells toward \glspl{tscm} when using membrane-bound IL15Ra and
IL15R\cite{Hurton2016}. 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.
@ -3424,7 +3472,7 @@ 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
\rmemh{} cells (~\cref{fig:nmr_cors}). Formate is a byproduct of the
one-carbon cycle implicated in promoting T cell activation\cite{RonHarel2016}.
Importantly, this cycle occurs between the cytosol and mitochondria of cells and
formate excreted\cite{Pietzke2020}. Mitochondrial biogenesis and function are
@ -3432,18 +3480,18 @@ shown necessary for memory cell persistence\cite{van_der_Windt_2012,
Vardhana2020}. 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 cells\cite{Lunt2011, Chang2013}. 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
activation\cite{Cao2014}. It may be that glycolytic cells dominate in the
culture at the early time points used for prediction, and higher lactate
reflects more cells.
In addition to formate, lactate was found as a putative \gls{cqa} of \ptmem{}
cells. Lactate is the end-product of aerobic glycolysis, characteristic of
highly proliferating cells and activated T cells\cite{Lunt2011, Chang2013}.
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 activation\cite{Cao2014}. 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
@ -3461,20 +3509,20 @@ reflects more cells.
% 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 glycolysis\cite{Cao2014}.
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 phenotypes\cite{Almeida2016,Wang_2012}. 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
primary carbon source (glucose) and essential amino acids (\gls{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
glycolysis\cite{Cao2014}. 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 phenotypes\cite{Almeida2016,Wang_2012}. 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, thus likely diluting some metabolic byproducts (such as formate,
lactate) and elevating depleted precursors (such as glucose and 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.