10
394 Org. Synth. 2012, 89, 394-403 Published on the Web 3/9/2012 © Organic Syntheses, Inc. Discussion Addendum For: Efficient Asymmetric Synthesis of N-tert-Butoxycarbonyl - Amino Acids using 4-tert-Butoxycarbonyl-5,6- Diphenylmorpholin-2-one: (R)-(N-tert-Butoxycarbonyl)allylglycine (4-Pentenoic acid, 2- [[(1,1-Dimethylethoxy)carbonyl]amino]-, (2R)-) t-BocN O O Ph Ph + I LiHMDS t-BocN O O Ph Ph H t-BocN O O Ph Ph H Li/NH 3 THF/EtOH t-BocHN O OH H THF Prepared by Ryan E. Looper 1 and Robert M. Williams.* 2 Original article: Williams, R. M.; Sinclair, P. J.; Demong, D. E. Org. Synth. 2003, 80, 31–37. The use of the diphenylmorpholinone template for the synthesis of optically enriched amino acid derivatives remains a highly selective, practical and predictable method. 3 While the chiral auxiliary is destroyed (by its eventual conversion to dibenzyl) and may not be ideally suited for synthetic applications on an industrial scale, it is a versatile tool for the preparation of research quantities of unusual or rare amino acid derivatives. The sense of stereo-induction with this template is generally very high and very predictable with new substituents being added to the -carbon anti- to the phenyl groups. Further the reactivity of this template is highly flexible. 4 The -carbon can react not only as an enolate, as described in the original Organic Syntheses preparation, but can also serve as an electrophile (via the iminium ion), radical, 1,3-dipole (azomethine ylide) 5,6,7 or can be converted to and used as the -phosphonate. It should also be noted that both antipodes of the template are commercially available, which permits the synthesis of both enantiomers of a given target with equal efficiency. Since the original report in Organic Syntheses on the preparation of (R)-(N-tert-butoxycarbonyl)allylglycine a number of groups have used this DOI:10.15227/orgsyn.089.0394

Discussion Addendum For: Efficient Asymmetric …practical and predictable method.3 While the chiral auxiliary is destroyed (by its eventual conversion to dibenzyl) and may not be

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Discussion Addendum For: Efficient Asymmetric …practical and predictable method.3 While the chiral auxiliary is destroyed (by its eventual conversion to dibenzyl) and may not be

394 Org. Synth. 2012, 89, 394-403

Published on the Web 3/9/2012

© Organic Syntheses, Inc.

Discussion Addendum For:

Efficient Asymmetric Synthesis of N-tert-Butoxycarbonyl -

Amino Acids using 4-tert-Butoxycarbonyl-5,6-

Diphenylmorpholin-2-one:

(R)-(N-tert-Butoxycarbonyl)allylglycine (4-Pentenoic acid, 2-

[[(1,1-Dimethylethoxy)carbonyl]amino]-, (2R)-)

t-BocN

O

O

Ph

Ph

+I

LiHMDS

t-BocN

O

O

Ph

Ph

H

t-BocN

O

O

Ph

Ph

H

Li/NH3

THF/EtOH

t-BocHN

O

OH

H

THF

Prepared by Ryan E. Looper1 and Robert M. Williams.*

2

Original article: Williams, R. M.; Sinclair, P. J.; Demong, D. E. Org. Synth.

2003, 80, 31–37.

The use of the diphenylmorpholinone template for the synthesis of

optically enriched amino acid derivatives remains a highly selective,

practical and predictable method.3 While the chiral auxiliary is destroyed (by

its eventual conversion to dibenzyl) and may not be ideally suited for

synthetic applications on an industrial scale, it is a versatile tool for the

preparation of research quantities of unusual or rare amino acid derivatives.

The sense of stereo-induction with this template is generally very high and

very predictable with new substituents being added to the -carbon anti- to

the phenyl groups. Further the reactivity of this template is highly flexible.4

The -carbon can react not only as an enolate, as described in the original

Organic Syntheses preparation, but can also serve as an electrophile (via the

iminium ion), radical, 1,3-dipole (azomethine ylide)5,6,7

or can be converted

to and used as the -phosphonate. It should also be noted that both antipodes

of the template are commercially available, which permits the synthesis of

both enantiomers of a given target with equal efficiency.

Since the original report in Organic Syntheses on the preparation of

(R)-(N-tert-butoxycarbonyl)allylglycine a number of groups have used this

DOI:10.15227/orgsyn.089.0394

Page 2: Discussion Addendum For: Efficient Asymmetric …practical and predictable method.3 While the chiral auxiliary is destroyed (by its eventual conversion to dibenzyl) and may not be

Org. Synth. 2012, 89, 394-403 395

allylglycine derivative for a variety of applications. Most often it is

incorporated as a metathesis precursor. For example, Nolen and co-workers

investigated the preparation of a more stable C-glycosyl serine mimic (5), to

be used in investigations of the O-glycosylation of serine residues which are

a ubiquitous post-translational modification for controlling biological

interactions (Fig. 1).8 The allylglycine derivative 1, derived from the

Organic Syntheses preparation, served as a cross-metathesis partner with

gluco-heptenitol. Thus treatment of 1 and 2 with Grubb’s 2nd

generation Ru-

catalyst gave the cyclization precursor 3 in 70% yield. Oxymercuration with

the pendent alcohol generated the C-glycoside 4 in high yield and as a single

diastereomer after reduction of the alkyl-mercurial species with Et3B and

NaBH4. The acetoxy derivative 5, poised for elaboration as a peptide

fragment, can be obtained after hydrogenolysis and acetylation.

BnO

OH

OBn

OBn

OH

BnO

OH

OBn

OBn

OH

CO2Me

NHBoc

O

CO2Me

NHBocBnOBnO

OBn

HO

O

CO2Me

NHBocAcOAcO

OAc

AcO

CO2Me

NHBoc

H H

15 mol% G2

CH2Cl2 (70%)

i. Hg(OTFA)2,

aq. KCl

ii. Et3B, NaBH4

(94%)

i. H2, Pd/C

ii. Ac2O, pyr.

1 2 3

4 5 Figure 1. Application of cross methathesis to allylglycine derivatives.

Cross-metathesis between an allylglycine derivative and tetradecene,

followed by hydrogenation, has also been used to create (R)-

aminoheptadecanoic acid (Fig. 2A). This long chain amino acid has found

use in the generation of truncated muraymicin analogues with increased cell

permeability.9 Ring closing metathesis (RCM) with two allylglycine units

has also been reported. This technology permitted the synthesis of 14-

membered macrocyclic peptides which were shown to be selective

antagonists of the d and m opioid receptors (Fig. 2B).10

Medium size rings

can also be generated through RCM with allylglycine derivatives. This has

found use in the development of Smac (second mitochondria-derived

activator of caspases) mimetics (Fig. 2C)11

and in the development of matrix

metalloproteinase (MMP) inhibitors inspired by the natural product cobactin

T (Fig. 2D).12

Page 3: Discussion Addendum For: Efficient Asymmetric …practical and predictable method.3 While the chiral auxiliary is destroyed (by its eventual conversion to dibenzyl) and may not be

396 Org. Synth. 2012, 89, 394-403

NNH

S

O O

O

Cl

OOH

cobactin T inspired MMP inhibitors

ON

NH

O

OHO2C

HN O

O

H2N

HO

HO

Me

HN

O

NH

O

H2N

NH

NH

NH

lipophilic muraymycin analogues

HO OH

metathesis

metathesis

NH

O

HN

HN

O

O

H2N

Ph

NH

HO2C

O

HO

metathesis

/μ selective opioid receptor antagonists

N

ONH

O

HN

MeMe

metathesis

NHO

Ph

H

Smac mimetics

A B

CD

Figure 2. Medicinally relevant small molecule derived from allylglycine

methathesis reactions.

Both (R) and (S)-(N-tert-butoxycarbonyl)allylglycine are now

commercially available, albeit generally 2-3 times more expensive than the

oxazinone template itself. As such, the oxazinone’s inherent applicability

might ultimately lie in the preparation of novel allylic amino acid

derivatives. Below we report on some recent examples in natural product

synthesis, medicinal chemistry, and the generation of unnatural amino acid

mimetics.

Funk and Greshock described the utility of their bromomethyl vinyl

ketone equivalent (6-bromomethyl-4H-1,3-dioxin, 6) for the synthesis of the

naturally occurring (2S,4R)-4-hydroxypipecolic acid (11) (Figure 3).13

Alkylation of the oxazinone with 6 gave 7 as a single diastereomer.

Thermolysis of this intermediate triggered a retrocycloaddition to reveal the

vinyl ketone 8, which underwent intramolecular Michael addition after

removal of the N-tert-butoxycarbonyl group with TMSOTf. Reduction of the

ketone group in 9 with borane gave 10 as a single diastereomer.

Hydrogenolysis of the auxiliary provided 11 in good yield and high optical

purity.

Page 4: Discussion Addendum For: Efficient Asymmetric …practical and predictable method.3 While the chiral auxiliary is destroyed (by its eventual conversion to dibenzyl) and may not be

Org. Synth. 2012, 89, 394-403 397

(S)

t-BocN

O(R)

O

Ph

Pht-BocN

O

O

Ph

Ph

O O

t-BocN

O

O

Ph

Ph

O

N

O

O

Ph

Ph

OH

HN

O

OH

OH

NaHMDS,

HMPA, - 78 oC

OO

Br

i. 150 oC,

PhMe, 3 h.

ii. TMSOTf, lutidine, MeOH

(59%, 2 steps)

(86%)

BH3, THF

- 78 oC, 20 min

(83%)

Pd(OH)2

H2 (50 psi)

(97%)

78

9

N

O

O

Ph

Ph

O 10

6

11

EtOAc

Figure 3. Allylic alkylation en route to (2S,4R)-4-hydroxypipecolic acid.

In 2004 we reported the use of the crotylglycine derivative 13 in the

synthesis of cylindrospermopsin,14

7-epicylindrospermopsin,15

and 7-

deoxycylindrospermopsin16

(Figure 4). Alkylation of the oxazinone with

crotyl iodide gave 12 in 92% yield and as a single diastereomer. Removal of

the auxiliary then gave (R)-(N-tert-butoxycarbonyl)crotylglycine in

reasonable yield and with high optical purity (> 99 : 1 e.r.). This was further

processed to the oxazinone-N-oxide 14, which underwent an intramolecular

[3+2]-cycloaddition to give the tricyclic isoxazolidine 15, thereby setting the

three contiguous stereocenters in the A ring of these natural products.17

The

key tricyclic intermediate 15 was then deployed in total syntheses of all

three naturally occurring cylindrospermopsin alkaloids, through initial

reductive cleavage of the N-O bond, differentiation of the two one-carbon

arms and elaboration.

t-BocN

O

O

Ph

Ph

I

NaHMDSt-BocN

O

O

Ph

Ph

H

Me

Li, NH3

THF, EtOH

(92%)

t-BocHN

O

OH

Me

H

(68%, > 99:1 e.r.)

N

O

Me

H

O

ON

O

OO

Me

H H200 oC

N NH

NH

OH

HN NH

O

O

Me

O3SO

N NH

NH

OH

HN NH

O

O

Me

O3SO

N NH

NH

HN NH

O

O

Me

O3SO

cylindrospermopsin 7-epicylindrospermopsin 7-deoxycylindrospermopsin

12 13

14 15

Me

THF

PhMe

A AA

Figure 4. Allylic alkyation to access the cylindrospermopsin alkaloids.

Page 5: Discussion Addendum For: Efficient Asymmetric …practical and predictable method.3 While the chiral auxiliary is destroyed (by its eventual conversion to dibenzyl) and may not be

398 Org. Synth. 2012, 89, 394-403

Researchers at Merck have utilized this methodology in the synthesis

of a novel series of potent and selective thrombin inhibitors (Figure 5).18

Alkylation of the oxazinone with methallyl bromide gave the intermediate

16 in acceptable yield. Cyclopropanation with diazomethane and Pd(OAc)2

then gave the pendent cyclopropane in 17, which survived dissolving metal

reduction of the auxiliary to give (R)-(N-tert-butoxycarbonyl)-

(methylcyclopropyl)propanoic acid (18). Incorporation of this fragment into

their common fluoro-proline scaffold gave the thrombin inhibitor 19 (Ki =

0.37 nM), which was nearly 1,000 fold selective for thrombin over trypsin.

t-BocN

O

O

Ph

Ph

t-BocN

O

O

Ph

Ph

Me

t-BocN

O

O

Ph

Ph

Me

t-BocHN

O

OH

Me

NaHMDS

THF, -78 oC

(46%)

(86%)

Li / NH3

THF , EtOH

(86%)

H

Cl

N

HN

O

N

N

N

O

NH2Me

F

Ki (thrombin) = 0.37 nM

Ki (trypsin) = 3300 nM

2xAPTT = 190 nM

H

H

H

16 17

18 19

MeBr

CH2N2

Pd(OAc)2

Figure 5. Allylic alkylation en route to thrombin inhibitors.

These types of reactions have been used in the preparation of other

medicinally relevant compounds as shown in Figure 6. Alkylation of the

oxazinone with an allylic phthalimide bearing a vinyl fluoride gave 20,

which has found use in the development of arginine mimetics as nitric oxide

synthase inhibitors (e.g. 21) (Fig. 6A).19

It should also be noted that the

allylated oxazinone originally described in the Organic Syntheses

preparation can undergo a second round of alkylation to generate a

quaternary -carbon with good selectivity, again with the second

electrophile approaching the enolate from the face opposite to the phenyl

rings. For example, alkylation with a pinacolatoborane reagent give the

intermediate 22, which has found application in the synthesis of arginase

inhibitors of the type 23 through oxidative cleavage of the allyl group and

reductive amination (Fig. 6B).20

A similar sequence can be carried out to

Page 6: Discussion Addendum For: Efficient Asymmetric …practical and predictable method.3 While the chiral auxiliary is destroyed (by its eventual conversion to dibenzyl) and may not be

Org. Synth. 2012, 89, 394-403 399

alkylate the allylated oxazinone with BOMCl giving 24 (Fig. 6C). Removal

of the auxiliary and benzyl ether provides (S)-(N-tert-butoxycarbonyl)-

(hydroxymethyl)-pentenoic acid (25). This intermediate has found utility in

the preparation of pyrrolothiazoles as inhibitors of dipeptidyl peptidase IV

(DPP-IV).21

t-Boc N

O

Ph

Ph

B

O

H2N

O

N

B

OH

HO

OH

R2

R1

arginase inhibitors

O

O

MeMe

Me

Me

t-BocN

O

Ph

Ph

HO

F

N

nitric oxide synthase inhibitors

O

O

H2N

O

HOH

F

NH

NHH2N

dipeptidyl peptidase IV (DPP-IV) inhibitors

t-BocN

O

Ph

Ph

O

O

Ph

t-BocN

O

OH

OHS

N

ONC

H

NH2

N

20 2122 23

2425 26

A B

C

Figure 6. Allylic alkylations to generate medicinally important small

molecules.

Another important use for allylglycine and its derivatives has been in

the generation of conformationally constrained -helical peptides. An early

report from our lab demonstrated that the tethered bis-allylglycine substrate

(27) underwent olefin metathesis to give an E/Z-mixture of macrocyclic

olefin products that upon hydrogenation provided the differentially protected

2,7-diaminosuberic acid derivative 28 (Figure 7).22

O

O

HN

O

O

PhO2C

tBOCHN1. Ru(cat)

2. H2 / Pdo tBOCHN

HO2C

CO2Ph

NH2

2827

Figure 7. Synthesis of a differentially protected 2,7-diaminosuberic acid

derivative.

Page 7: Discussion Addendum For: Efficient Asymmetric …practical and predictable method.3 While the chiral auxiliary is destroyed (by its eventual conversion to dibenzyl) and may not be

400 Org. Synth. 2012, 89, 394-403

The conceptually related intramolecular metathesis of allylglycine-

containing peptides, has been pioneered by Verdine to make

conformationally-constrained -helical peptides which have been named

“stapled peptides”.23

As shown in Figure 8A, the oxazinone template was

used to synthesize both enantiomeric series of -methyl- -alkenyl amino

acids. The , -disubstitution increases helicity, while a variation in tether

length allowed the examination of chemical yield for the metathesis reaction.

These amino acids were incorporated into model peptides at the i and i+4

positions. Metathesis between the two alkene termini successfully “stapled”

the peptides together. The shortest metathesis reaction tolerated was that

between the allylglycine derivative (S-11) and hexenyl-substituted amino

acid (S-14) as shown in Figure 8B. The opposite R,R enantiomeric series

was also successful while the R,S series failed to metathesize. An increase in

tether length increased metathesis efficiency leading to reactions with >98%

conversion. Depending on the length of the tether, these peptides possessed

greater helicity and resistance to proteolysis.

The original article detailing the preparation of (R)-(N-tert-

butoxycarbonyl)allylglycine provides a useful route to prepare this

commonly used and now commercially available amino acid for the

synthesis of more complex unnatural amino acids and elaborated peptides.

Perhaps more impressive is the utility of the oxazinone to prepare more

ornate allylic-amino acid derivatives for use in natural products and

medicinal chemistry. Finally, the ease of preparation of the oxazinones24

(also commercially available) allows for the synthesis of stable isotopomers

of allylglycine, which can be introduced into the oxazinone template25

via

bromoacetate or via the allyl iodide fragment. The incorporation of

deuterium, 13

C, and 15

N into this system can thus be accomplished in several

reasonably economical ways and can be of use in a variety of applications.

Page 8: Discussion Addendum For: Efficient Asymmetric …practical and predictable method.3 While the chiral auxiliary is destroyed (by its eventual conversion to dibenzyl) and may not be

Org. Synth. 2012, 89, 394-403 401

HN

Fmoc OH

O

Me

HN

Fmoc OH

O

Me

17% conversionby methathesis

R-11

R-14

HN

Fmoc OH

O

Me

S-11

HN

Fmoc OH

O

Me

S-14

68% conversionby methathesis

B

Si,i+4S(7) Ri,i+4R(7)

N

OPh

Ph

tBoc O

N

OPh

Ph

tBoc O

HN

Fmoc OH

O

Me

Men

HN

Fmoc OH

O

Me

N

OPh

Ph

tBoc O

n

N

OPh

Ph

tBoc O

1. KMDS, MeI

2. KHMDSI

n

1. Na, NH3

2. TFA, then

FmocOSu,

Na2CO3

Me

1. KMDS, MeI

2. KHMDSI

n n

n

1. Na, NH3

2. TFA, then

FmocOSu,

Na2CO3

(n=1,2,3,4,6)

(n=1,3,4)

A

Figure 8. Synthesis of , -disubstituted amino acids and their incorporation

into stapled peptides. Adapted with permission from J. Am. Chem. Soc.

2000, 122, 5891–5892, Copyright (2000) American Chemical Society.

1. Department Of Chemistry, University of Utah, Salt Lake City, UT

84112

2. Department of Chemistry, Colorado State University, Fort Collins, CO,

80523

Page 9: Discussion Addendum For: Efficient Asymmetric …practical and predictable method.3 While the chiral auxiliary is destroyed (by its eventual conversion to dibenzyl) and may not be

402 Org. Synth. 2012, 89, 394-403

3. Williams, R. M.; Im, M. N. J. Am. Chem. Soc. 1991, 113, 9276–9286.

4. Reviews: Asymmetric Synthesis of a-Amino Acids. Williams, R.M.,

Advances in Asymmetric Synthesis, JAI Press 1995, Volume 1 (pp 45–

94) A. Hassner, Ed.

5. Sebahar, P.; Williams, R. M. J. Am. Chem. Soc. 2000, 122, 5666–5667.

6. Sebahar, P. R.; Osada, H.; Usui T.; Williams, R. M. Tetrahedron 2002

58, 6311–6322.

7. Lo, M. M.-C.; Neumann, C. S.; Nagayama, S.; Perlstein, E. O.;

Schreiber, S. L. J. Am. Chem. Soc. 2004, 126, 16077–16086.

8. Nolen, E. G.; Kurish, A. J.; Potter, J. M.; Donahue, L. A.; Orlando, M.

D. Org. Lett. 2005, 7, 3383–3386.

9. Tanino, T.; Al-Dabbagh, B.; Mengin-Lecreulx, D.; Bouhss, A.; Oyama,

H.; Ichikawa, S.; Matsuda, A. J. Med. Chem. 2011, 54, 8421–8439.

10. Mollica, A.; Guardiani, G.; Davis, P.; Ma, S.-W.; Porreca, F.; Lai, J.;

Mannina, L.; Sobolev, A. P.; Hruby, V. J. J. Med. Chem. 2007,

50, 3138–3142.

11. Sun, W.; Nikolovska-Coleska, Z.; Qin, D.; Sun, H.; Yang, C.-Y.; Bai,

L.; Qiu, S.; Wang, Y.; Ma, D.; Wang, S. J. Med. Chem. 2009, 52, 593–

596.

12. Wilson, L. J.; Wang, B.; Yang, S.-M.; Scannevin, R. H.; Burke, S. L.;

Karnachi, P.; Rhodes, K. J.; Murray, W. V. Bioorg. Med. Chem.

Lett. 2011, 21, 6485–6490.

13. Greshock, T. J.; Funk, R. L. J. Am. Chem. Soc. 2002, 124, 754–755.

14. Looper, R. E.; Runnegar, M. T. C.; Williams, R. M. Tetrahedron 2006,

62, 4549–4562.

15. Looper, R. E.; Williams, R. M. Angew. Chem. Int. Ed. 2004, 43, 2930–

2933.

16. Looper, R. E.; Runnegar, M. T. C.; Williams, R. M. Angew. Chem. Int.

Ed. 2005, 44, 3879–3881.

17. Looper, R. E.; Williams, R. M. Tetrahedron Lett. 2001, 42, 769–771.

18. Staas, D. D.; Savage, K. L.; Sherman, V. L.; Shimp, H. L.; Lyle, T. A.;

Tran, L. O.; Wiscount, C. M.; McMasters, D. R.; Sanderson, P. E. J.;

Williams, P. D.; Lucas, B. J.; Krueger, J. A.; Lewis, S. D.; White, R. B.;

Yu, S.; Wong, B. K.; Kochansky, C. J.; Anari, M. R.; Yand, Y.; Vacca,

J. P. Bioorg. Med. Chem. 2006, 14, 6900–6916.

19. Hansen, D. W., Jr.; Webber, R. K.; Awasthi, A. K.; Manning, P. T.;

Sikorski, J. A. PCTInt. Appl. (2001), WO 2001079156 A1 20011025.

Page 10: Discussion Addendum For: Efficient Asymmetric …practical and predictable method.3 While the chiral auxiliary is destroyed (by its eventual conversion to dibenzyl) and may not be

Org. Synth. 2012, 89, 394-403 403

20. Van Zandt, M.; Golebiowski, A.; Ji, M. K.; Whitehouse, D.; Ryder, T.;

Beckett, P. PCT Int. Appl. (2011), WO 2011133653 A1 20111027.

21. Wagner, H.; Schoenafinger, K.; Jaehne, G.; Gaul, H.; Buning, C.;

Tschank, G.; Werner, U. PCT Int. Appl. (2005), WO 2005012312 A1

20050210.

22. Williams, R. M.; Liu, J. J. Org. Chem. 1998, 63, 2130–2132.

23. Schafmeister, C. E.; Po, J.; Verdine, G. L. J. Am. Chem. Soc. 2000, 122,

5891–5892.

24. Williams, R. M.; Sinclair, P. J.; DeMong, D.; Chen, D.; Zhai, D., Org.

Synth. 2003, 80, 18–30.

25. Aoyagi, Y.; Iijima, A.; Williams, R. M., J. Org. Chem. 2001, 66, 8010–

8014.

Robert M. Williams was born in New York in 1953 and

attended Syracuse University where he received the B.A.

degree in Chemistry in 1975. He obtained the Ph.D. degree in

1979 at MIT (W. H. Rastetter) and was a post-doctoral fellow

at Harvard (1979-1980; R. B. Woodward/Yoshito Kishi). He

joined Colorado State University in 1980 and was named a

University Distinguished Professor in 2002. His

interdisciplinary research program (over 280 publications) at

the chemistry-biology interface is focused on the total synthesis

of biomedically significant natural products, biosynthesis of

secondary metabolites, studies on antitumor drug-DNA

interactions, HDAC inhibitors, amino acids and peptides.

Ryan E. Looper was born in Banbury, England in 1976. He

came to the U.S. to attend Western Washington University

where he earned a B.S. degree in Chemistry in 1998 and an

M.S. degree in 1999 under the guidance of J. R. Vyvyan. He

obtained his Ph.D. degree in 2004 at Colorado State University

in the laboratories of Prof. R.M. Williams. After a NIH post-

doctoral fellowship at Harvard University with Prof. S.L.

Schreiber, he began his independent career at the University of

Utah in 2007.